51
|
Ben Musa R, Cornelius-Green J, Hasser EM, Cummings KJ. The effect of orexin on the hypoxic ventilatory response of female rats is greatest in the active phase during diestrus. J Appl Physiol (1985) 2023; 134:638-648. [PMID: 36656978 PMCID: PMC10010922 DOI: 10.1152/japplphysiol.00661.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023] Open
Abstract
We recently showed that in male rats, orexin contributes to the hypoxic ventilatory response (HVR), with a stronger effect in the active phase. The effect of orexin on the HVR in females has not been investigated. As estrogen can inhibit orexin neurons, here we hypothesized that orexin neurons are activated by hypoxia and facilitate the HVR only in diestrus, when estrogen is low. We exposed female rats (n = 10) to near-isocapnic hypoxia ([Formula: see text] from 0.21 to 0.09) over ∼5 min, after vehicle and again after suvorexant (a dual OxR antagonist; 20 mg/kg ip), with ventilation measured using whole body plethysmography. Each rat was tested in proestrus or estrus (p/estrus), and again in diestrus, during both inactive and active phases. We also performed immunohistochemistry (IHC) to determine the proportion of orexin neurons activated by acute hypoxia during diestrus (n = 6) or proestrus/estrus (n = 6) in the active phase. In the inactive phase, the HVR was unaffected by OxR blockade, irrespective of estrus stage. In the active phase, the effect of OxR blockade depended on stage: the slope of the HVR was significantly reduced by OxR blockade only during diestrus. IHC revealed that hypoxia activated more orexin neurons during diestrus compared with p/estrus. We conclude that in females, orexin neurons are activated by hypoxia and contribute to the HVR only in diestrus when estrogen levels are low. Stage of the estrus cycle should be considered when examining the physiological function of orexin neurons in females.NEW & NOTEWORTHY We previously showed that orexin facilitates the hypoxic ventilatory response (HVR) of adult male rats during the active phase. Others have shown that estrogen inhibits orexin neurons. Here we show that orexin neurons are activated by hypoxia and facilitate the HVR of adult female rats during the active phase, but only in diestrus. These data suggest that orexin neurons facilitate the HVR in females when they are free from the inhibitory effects of estrogen.
Collapse
Affiliation(s)
- Ruwaida Ben Musa
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Jennifer Cornelius-Green
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Eileen M Hasser
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Kevin J Cummings
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
52
|
Rijsketic DR, Casey AB, Barbosa DA, Zhang X, Hietamies TM, Ramirez-Ovalle G, Pomrenze M, Halpern CH, Williams LM, Malenka RC, Heifets BD. UNRAVELing the synergistic effects of psilocybin and environment on brain-wide immediate early gene expression in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.19.528997. [PMID: 36865251 PMCID: PMC9980055 DOI: 10.1101/2023.02.19.528997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The effects of context on the subjective experience of serotonergic psychedelics have not been fully examined in human neuroimaging studies, partly due to limitations of the imaging environment. Here, we administered saline or psilocybin to mice in their home cage or an enriched environment, immunofluorescently-labeled brain-wide c-Fos, and imaged cleared tissue with light sheet microscopy to examine the impact of context on psilocybin-elicited neural activity at cellular resolution. Voxel-wise analysis of c-Fos-immunofluorescence revealed differential neural activity, which we validated with c-Fos + cell density measurements. Psilocybin increased c-Fos expression in the neocortex, caudoputamen, central amygdala, and parasubthalamic nucleus and decreased c-Fos in the hypothalamus, cortical amygdala, striatum, and pallidum. Main effects of context and psilocybin-treatment were robust, widespread, and spatially distinct, whereas interactions were surprisingly sparse.
Collapse
Affiliation(s)
- Daniel Ryskamp Rijsketic
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Austen B. Casey
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Daniel A.N. Barbosa
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xue Zhang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Tuuli M. Hietamies
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Grecia Ramirez-Ovalle
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Matthew Pomrenze
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
- Nancy Pritzker Laboratory, Stanford University, Stanford, CA 94305, USA
| | - Casey H. Halpern
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leanne M. Williams
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
- Sierra-Pacific Mental Illness Research, Education, and Clinical Center (MIRECC) Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Robert C. Malenka
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
- Nancy Pritzker Laboratory, Stanford University, Stanford, CA 94305, USA
| | - Boris D. Heifets
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
53
|
Iizuka T, Mori C, Okanoya K. Song-related brain auditory activity in Bengalese finches as examined by immediate early gene expressions: Comparison of arousal states and the correlational analyses between brain regions. Neurosci Res 2023:S0168-0102(23)00024-X. [PMID: 36740096 DOI: 10.1016/j.neures.2023.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/11/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023]
Abstract
Songbirds use auditory feedback to memorize a tutor song in juveniles and to maintain it in adults. In Bengalese finches, electrophysiological studies showed the auditory responses in the premotor area HVC remained active regardless of asleep/awake status, in contrast to auditory gating phenomenon identified in zebra finches. We investigated the correlations in auditory activity between the brain regions and differences in the activity during wakefulness and sleeping in Bengalese finches. We used the immediate early gene egr-1 as a marker of neural activity that can detect regions responding to auditory stimuli in the whole brain. Results showed that auditory response, as measured by egr-1 expression to the bird's own song while sleeping and awake, was similar in HVC and NCM. Higher activity during awake than sleep was found only in the lower auditory area MLd. Analyses showed egr-1 expressions between brain regions induced by the bird's own song playback in awake/sleep conditions, suggesting that auditory information correlated with the inter part, not the outer part, of MLd with the higher song-related regions. Furthermore, the sleep condition suppressed the spontaneous activity, but not the song-induced activity in Area X. Altogether, this study presents a new attempt to explore the auditory-motor network using a molecular tool to map neurons of the nearly whole brain.
Collapse
Affiliation(s)
- Takafumi Iizuka
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan; Advanced Comprehensive Research Organization, Teikyo University, 2-21-1 Kaga, Itabashi-ku, Tokyo 173-0003, Japan
| | - Chihiro Mori
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan; Department of Molecular Biology, Faculty of Pharmaceutical Science, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Kazuo Okanoya
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan; Advanced Comprehensive Research Organization, Teikyo University, 2-21-1 Kaga, Itabashi-ku, Tokyo 173-0003, Japan.
| |
Collapse
|
54
|
Neural mechanism underlying depressive-like state associated with social status loss. Cell 2023; 186:560-576.e17. [PMID: 36693374 DOI: 10.1016/j.cell.2022.12.033] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 10/13/2022] [Accepted: 12/20/2022] [Indexed: 01/25/2023]
Abstract
Downward social mobility is a well-known mental risk factor for depression, but its neural mechanism remains elusive. Here, by forcing mice to lose against their subordinates in a non-violent social contest, we lower their social ranks stably and induce depressive-like behaviors. These rank-decline-associated depressive-like behaviors can be reversed by regaining social status. In vivo fiber photometry and single-unit electrophysiological recording show that forced loss, but not natural loss, generates negative reward prediction error (RPE). Through the lateral hypothalamus, the RPE strongly activates the brain's anti-reward center, the lateral habenula (LHb). LHb activation inhibits the medial prefrontal cortex (mPFC) that controls social competitiveness and reinforces retreats in contests. These results reveal the core neural mechanisms mutually promoting social status loss and depressive behaviors. The intertwined neuronal signaling controlling mPFC and LHb activities provides a mechanistic foundation for the crosstalk between social mobility and psychological disorder, unveiling a promising target for intervention.
Collapse
|
55
|
Davoudian PA, Shao LX, Kwan AC. Shared and Distinct Brain Regions Targeted for Immediate Early Gene Expression by Ketamine and Psilocybin. ACS Chem Neurosci 2023; 14:468-480. [PMID: 36630309 PMCID: PMC9898239 DOI: 10.1021/acschemneuro.2c00637] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Psilocybin is a psychedelic with therapeutic potential. While there is growing evidence that psilocybin exerts its beneficial effects through enhancing neural plasticity, the exact brain regions involved are not completely understood. Determining the impact of psilocybin on plasticity-related gene expression throughout the brain can broaden our understanding of the neural circuits involved in psychedelic-evoked neural plasticity. In this study, whole-brain serial two-photon microscopy and light sheet microscopy were employed to map the expression of the immediate early gene, c-Fos, in male and female mice. The drug-induced c-Fos expression following psilocybin administration was compared to that of subanesthetic ketamine and saline control. Psilocybin and ketamine produced acutely comparable elevations in c-Fos expression in numerous brain regions, including anterior cingulate cortex, locus coeruleus, primary visual cortex, central and basolateral amygdala, medial and lateral habenula, and claustrum. Select regions exhibited drug-preferential differences, such as dorsal raphe and insular cortex for psilocybin and the CA1 subfield of hippocampus for ketamine. To gain insights into the contributions of receptors and cell types, the c-Fos expression maps were related to brain-wide in situ hybridization data. The transcript analyses showed that the endogenous levels of Grin2a and Grin2b predict whether a cortical region is sensitive to drug-evoked neural plasticity for both ketamine and psilocybin. Collectively, the systematic mapping approach produced an unbiased list of brain regions impacted by psilocybin and ketamine. The data are a resource that highlights previously underappreciated regions for future investigations. Furthermore, the robust relationships between drug-evoked c-Fos expression and endogenous transcript distributions suggest glutamatergic receptors as a potential convergent target for how psilocybin and ketamine produce their rapid-acting and long-lasting therapeutic effects.
Collapse
Affiliation(s)
- Pasha A. Davoudian
- Medical Scientist Training Program, Yale University School of Medicine, New Haven, Connecticut, 06511, USA
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, Connecticut, 06511, USA
| | - Ling-Xiao Shao
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, 06511, USA
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, 14853, USA
| | - Alex C. Kwan
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, 06511, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, 06511, USA
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, 14853, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, New York, 10065, USA
| |
Collapse
|
56
|
Yoshimura Y, Nakamura K, Seno M, Mochizuki M, Kawai K, Koba S, Watanabe T. Generation of c-Fos knockout rats, and observation of their phenotype. Exp Anim 2023; 72:95-102. [PMID: 36216550 PMCID: PMC9978135 DOI: 10.1538/expanim.22-0077] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
c-Fos is a useful marker gene of neuron activation for neuroscience and physiology research. The mechanism and function of neural networks have been elucidated using c-Fos reporter knock-in (KI) mice, but the small size of the mice makes it difficult to perform surgical procedures on specific brain regions. On the other hand, there is a large amount of accumulated data on behavioral studies using rats. Thus, the generation of c-Fos reporter rat is expected, but it is difficult to generate gene-modified rats. Furthermore, c-Fos gene abnormality is expected to be severe in rats, as shown in homozygous of c-Fos knockout (KO) mouse, but such analysis has rarely been performed and is not certain. This study generated c-Fos-deficient rats using CRISPR/Cas, with 1067 bp deletion including exon 1 of the c-Fos gene. Homozygous c-Fos KO rats had growth latency and the same tooth and bone abnormality as homozygous c-Fos KO mice but not heterozygous c-Fos KO rats. Therefore, the c-Fos gene in rats is expected to have the same function as that in mice, and the generation of c-Fos reporter KI rats is further anticipated.
Collapse
Affiliation(s)
- Yuki Yoshimura
- Division of Integrative Physiology, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Kazuomi Nakamura
- Advanced Medicine Innovation and Clinical Research Center, Tottori University Hospital, 36-1 Nishi-cho, Yonago, Tottori 683-8504, Japan,Advanced Medicine & Translational Research Center, Organization for Research Initiative and Promotion, Tottori University, 86 Nishi-cho, Yonago, Tottori
683-8503, Japan
| | - Misako Seno
- Advanced Medicine & Translational Research Center, Organization for Research Initiative and Promotion, Tottori University, 86 Nishi-cho, Yonago, Tottori
683-8503, Japan
| | - Misa Mochizuki
- Pathology Center, Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Kenji Kawai
- Pathology Center, Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Satoshi Koba
- Division of Integrative Physiology, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Tatsuo Watanabe
- Division of Integrative Physiology, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| |
Collapse
|
57
|
Leonardi CEP, Carrasco RA, Dias FCF, Zwiefelhofer EM, Adams GP, Singh J. Mechanism of LH release after peripheral administration of kisspeptin in cattle. PLoS One 2022; 17:e0278564. [PMID: 36459509 PMCID: PMC9718405 DOI: 10.1371/journal.pone.0278564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 11/18/2022] [Indexed: 12/04/2022] Open
Abstract
Kisspeptin modulates GnRH secretion in mammals and peripheral administration of 10-amino acid fragment of kisspeptin (Kp10) induces LH release and ovulation in cattle. Experiments were done to determine if iv administration of kisspeptin will activate GnRH neurons (i.e., after crossing the blood-brain barrier) and if pre-treatment with a GnRH receptor blocker will alter kisspeptin-induced LH release (from gonadotrophs) and ovulation. In Experiment 1, cows (n = 3 per group) were given human-Kisspeptin10 (hKp10; 3 x 15 mg iv at 60-min intervals) or normal saline and euthanized 150 min after treatment was initiated. Every 20th free-floating section (50 μm thickness) from the preoptic area to hypothalamus was double immunostained to colocalize GnRH- (DAB) and activated neurons (cFOS; Nickel-DAB). Kisspeptin induced plasma LH release from 15 to 150 min (P = 0.01) but the proportion of activated GnRH neurons did not differ between groups (5.8% and 3.5%, respectively; P = 0.11). Immunogold electron microscopy detected close contacts between kisspeptin fibers and GnRH terminals in the median eminence. In Experiment 2, pubertal heifers (n = 5 per group) were treated with 1) hKp10 iv, 2) Cetrorelix (GnRH antagonist; im) + hKp10 iv or 3) saline on Day 6 of the follicular wave under low-progesterone condition. A rise in plasma LH concentration was detected from 15 to 240 min in the hKp10 group but not in cetrorelix or control group (P<0.001). Ovulations were detected only in the hKp10 group (4/5; P = 0.02). Cetrorelix treatment was associated with regression of the preovulatory dominant follicle and emergence of a new follicular wave 3.4±0.75 days after the treatment in all five heifers. Results support the hypothesis that the effect of peripheral kisspeptin is mediated downstream of GnRH synthesis and does not involve GnRH-independent LH release from gonadotrophs. Peripheral kisspeptin may release pre-synthesized GnRH from the nerve terminals in areas outside the blood-brain barrier.
Collapse
Affiliation(s)
- Carlos E. P. Leonardi
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Rodrigo A. Carrasco
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Fernanda C. F. Dias
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Eric M. Zwiefelhofer
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Gregg P. Adams
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Jaswant Singh
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
- * E-mail:
| |
Collapse
|
58
|
Beacher NJ, Washington KA, Zhang Y, Li Y, Lin DT. GRIN lens applications for studying neurobiology of substance use disorder. ADDICTION NEUROSCIENCE 2022; 4:100049. [PMID: 36531187 PMCID: PMC9757736 DOI: 10.1016/j.addicn.2022.100049] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Substance use disorder (SUD) is associated with severe health and social consequences. Continued drug use results in alterations of circuits within the mesolimbic dopamine system. It is critical to observe longitudinal impacts of SUD on neural activity in vivo to identify SUD predispositions, develop pharmaceuticals to prevent overdose, and help people suffering from SUD. However, implicated SUD associated areas are buried in deep brain which makes in vivo observation of neural activity challenging. The gradient index (GRIN) lens can probe these regions in mice and rats. In this short communications review, we will discuss how the GRIN lens can be coupled with other technologies such as miniaturized microscopes, fiberscopes, fMRI, and optogenetics to fully explore in vivo SUD research. Particularly, GRIN lens allows in vivo observation of deep brain regions implicated in SUD, differentiation of genetically distinct neurons, examination of individual cells longitudinally, correlation of neuronal patters with SUD behavior, and manipulation of neural circuits.
Collapse
Affiliation(s)
- Nicholas James Beacher
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Kayden Alecsandre Washington
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Yan Zhang
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Yun Li
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, United States
| | - Da-Ting Lin
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
59
|
Calderazzo S, Covert M, Alba DD, Bowley BE, Pessina MA, Rosene DL, Buller B, Medalla M, Moore TL. Neural recovery after cortical injury: Effects of MSC derived extracellular vesicles on motor circuit remodeling in rhesus monkeys. IBRO Neurosci Rep 2022; 13:243-254. [PMID: 36590089 PMCID: PMC9795302 DOI: 10.1016/j.ibneur.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 07/01/2022] [Accepted: 08/07/2022] [Indexed: 01/04/2023] Open
Abstract
Reorganization of motor circuits in the cortex and corticospinal tract are thought to underlie functional recovery after cortical injury, but the mechanisms of neural plasticity that could be therapeutic targets remain unclear. Recent work from our group have shown that systemic treatment with mesenchymal stem cell derived (MSCd) extracellular vesicles (EVs) administered after cortical damage to the primary motor cortex (M1) of rhesus monkeys resulted in a robust recovery of fine motor function and reduced chronic inflammation. Here, we used immunohistochemistry for cfos, an activity-dependent intermediate early gene, to label task-related neurons in the surviving primary motor and premotor cortices, and markers of axonal and synaptic plasticity in the spinal cord. Compared to vehicle, EV treatment was associated with a greater density of cfos+ pyramidal neurons in the deep layers of M1, greater density of cfos+ inhibitory interneurons in premotor areas, and lower density of synapses on MAP2+ lower motor neurons in the cervical spinal cord. These data suggest that the anti-inflammatory effects of EVs may reduce injury-related upper motor neuron damage and hyperexcitability, as well as aberrant compensatory re-organization in the cervical spinal cord to improve motor function.
Collapse
Key Words
- CB, Calbindin
- CR, Calretinin
- CSC, Cervical Spinal Cord
- Circuit Remodeling
- Cortical Injury
- DH, Dorsal Horn
- EVs, Extracellular Vesicles
- Extracellular Vesicles
- Ischemia
- LCST, Lateral Corticospinal Tract
- M1, Primary Motor Cortex
- MAP2, Microtubule Associated Protein 2
- MSCd, Mesenchymal Stem Cell derived
- Motor Cortex
- NHP, Non-Human Primate
- PV, Parvalbumin
- Plasticity
- ROS, Reactive Oxygen Species
- SYN, Synaptophysin
- Stem Cell-Based Treatments
- VH, Ventral Horn
- dPMC, dorsal Premotor Cortex
- miRNA, Micro RNA
- periM1, Perilesional Primary Motor Cortex
Collapse
Affiliation(s)
| | | | | | | | | | - Douglas L. Rosene
- Anatomy and Neurobiology Dept, BUSM, USA
- Center for Systems Neuroscience, BU, USA
| | | | - Maria Medalla
- Anatomy and Neurobiology Dept, BUSM, USA
- Center for Systems Neuroscience, BU, USA
| | - Tara L. Moore
- Anatomy and Neurobiology Dept, BUSM, USA
- Center for Systems Neuroscience, BU, USA
| |
Collapse
|
60
|
Guo W, Fan S, Xiao D, He C, Guan M, Xiong W. A midbrain-reticulotegmental circuit underlies exaggerated startle under fear emotions. Mol Psychiatry 2022; 27:4881-4892. [PMID: 36117214 DOI: 10.1038/s41380-022-01782-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 01/19/2023]
Abstract
Exaggerated startle has been recognized as a core hyperarousal symptom of multiple fear-related anxiety disorders, such as post-traumatic stress disorder (PTSD) and panic disorder. However, the mechanisms driving this symptom are poorly understood. Here we reveal a neural projection from dorsal raphe nucleus (DRN) to a startle-controlling center reticulotegmental nucleus (RtTg) that mediates enhanced startle response under fear condition. Within RtTg, we identify an inhibitory microcircuit comprising GABAergic neurons in pericentral RtTg (RtTgP) and glutamatergic neurons in central RtTg (RtTgC). Inhibition of this RtTgP-RtTgC microcircuit leads to elevated startle amplitudes. Furthermore, we demonstrate that the conditioned fear-activated DRN 5-HTergic neurons send inhibitory projections to RtTgP GABAergic neurons, which in turn upregulate neuronal activities of RtTgC glutamatergic neurons. Chemogenetic activation of the DRN-RtTgP projections mimics the increased startle response under fear emotions. Moreover, conditional deletion of 5-HT1B receptor from RtTgP GABAergic neurons largely reverses the exaggeration of startle during conditioned fear. Thus, our study establishes the disinhibitory DRN-RtTgP-RtTgC circuit as a critical mechanism underlying exaggerated startle under fear emotions, and provides 5-HT1B receptor as a potential therapeutic target for treating hyperarousal symptom in fear-associated psychiatric disorders.
Collapse
Affiliation(s)
- Weiwei Guo
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Sijia Fan
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Dan Xiao
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Chen He
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Mengyuan Guan
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Wei Xiong
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China. .,Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230088, China. .,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
61
|
CO 2 exposure enhances Fos expression in hypothalamic neurons in rats during the light and dark phases of the diurnal cycle. Brain Struct Funct 2022; 227:2667-2679. [PMID: 36109371 DOI: 10.1007/s00429-022-02562-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 08/29/2022] [Indexed: 12/30/2022]
Abstract
Orexinergic (OX) neurons in the lateral hypothalamus (LH), perifornical area (PFA) and dorsomedial hypothalamus (DMH) play a role in the hypercapnic ventilatory response, presumably through direct inputs to central pattern generator sites and/or through interactions with other chemosensitive regions. OX neurons can produce and release orexins, excitatory neuropeptides involved in many functions, including physiological responses to changes in CO2/pH. Thus, in the present study, we tested the hypothesis that different nuclei (LH, PFA and DMH) where the orexinergic neurons are located, show distinct activation by CO2 during the light-dark cycle phases. For this purpose, we evaluated the Fos and OXA expression by immunohistochemistry to identify neurons that co-localize Fos + OXA in the LH, LPeF, MPeF and DMH in the light-inactive and dark-active phase in Wistar rats subjected to 3 h of normocapnia or hypercapnia (7% CO2). Quantitative analyses of immunoreactive neurons show that hypercapnia caused an increase in the number of neurons expressing Fos in the LH, LPeF, MPeF and DMH in the light and dark phases. In addition, the number of Fos + OXA neurons increased in the LPeF and DMH independently of the phases of the diurnal cycle; whereas in the MPeF, this increase was observed exclusively in the light phase. Thus, we suggest that OX neurons are selectively activated by hypercapnia throughout the diurnal cycle, reinforcing the differential role of nuclei in the hypothalamus during central chemosensitivity.
Collapse
|
62
|
Pan L, Zhang X, Gao Q. Histatin-1 alleviates high-glucose injury to skin keratinocytes through MAPK signaling pathway. J Cosmet Dermatol 2022; 21:6281-6291. [PMID: 35819887 DOI: 10.1111/jocd.15235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/05/2022] [Accepted: 07/08/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Damage to keratinocytes and other skin cells in a high-glucose environment has been proven to be an important reason for the poor wound healing ability of chronic diabetes mellitus. Histatin-1 has been preliminarily proven to stimulate the wound healing process of the oral and non-oral mucosa and has been found to be related to the activation of extracellular signal-regulated kinase (ERK). AIM OF THE STUDY The purpose of this study was to investigate the effect of histatin-1 on high-glucose-injured keratinocytes and the role of the Ras-Raf-MEK-ERK signaling pathway on the effect of histatin-1 to improve diabetic wound healing. METHODS A human keratinocyte model damaged by high glucose was constructed, cell proliferation was detected by the Cell Counting Kit-8 assay, and cell apoptosis was detected by flow cytometry. The expression level of 8-hydroxy-2'-deoxyguanosine (8-OHdG) was detected by ELISA, and the mitogen-activated protein kinase (MAPK) signaling pathway protein expression level was detected by Western blot. C-fos mRNA expression was detected by real-time PCR. RESULTS The results indicated that histatin-1 promoted proliferation and reduced the rate of apoptosis and 8-OHdG content in keratinocytes with high-glucose injury. In addition, histatin-1 down-regulated MEK phosphorylation in keratinocytes with high-glucose injury. However, with the extension of the intervention, the effect of histatin-1 on c-fos mRNA expression was different. At the early stage of high-glucose injury (12 h), the expression of c-fos mRNA was not increased in high-glucose-injured keratinocytes treated with histatin-1 but then c-fos mRNA expression was gradually upregulated. CONCLUSION Histatin-1 could alleviate keratinocyte injury caused by high glucose levels and promoted wound healing in vitro. In addition, histatin-1 could exert anti-apoptotic and antioxidant damage effects under high-glucose injury states. These effects of histatin-1 may be related to its regulation of the MAPK signaling pathway. Therefore, these findings provide an essential theoretical basis for histatin-1 to become a safe and effective new peptide biological agent to promote wound healing in patients with diabetes.
Collapse
Affiliation(s)
- Li Pan
- Department of Cardiopulmonary Bypass, Lanzhou University Second Hospital, Lanzhou, China
| | - Xuanfen Zhang
- Department of Plastic Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Qiong Gao
- Department of Plastic Surgery, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
63
|
Oestrogen-dependent hypothalamic oxytocin expression with changes in feeding and body weight in female rats. Commun Biol 2022; 5:912. [PMID: 36064966 PMCID: PMC9445083 DOI: 10.1038/s42003-022-03889-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 08/25/2022] [Indexed: 11/08/2022] Open
Abstract
Oxytocin (OXT) is produced in the hypothalamic nuclei and secreted into systemic circulation from the posterior pituitary gland. In the central nervous system, OXT regulates behaviours including maternal and feeding behaviours. Our aim is to evaluate whether oestrogen regulates hypothalamic OXT dynamics. Herein, we provide the first evidence that OXT dynamics in the hypothalamus vary with sex and that oestrogen may modulate dynamic changes in OXT levels, using OXT-mRFP1 transgenic rats. The fluorescence intensity of OXT-mRFP1 and expression of the OXT and mRFP1 genes in the hypothalamic nuclei is highest during the oestrus stage in female rats and decreased significantly in ovariectomised rats. Oestrogen replacement caused significant increases in fluorescence intensity and gene expression in a dose-related manner. This is also demonstrated in the rats' feeding behaviour and hypothalamic Fos neurons using cholecystokinin-8 and immunohistochemistry. Hypothalamic OXT expression is oestrogen-dependent and can be enhanced centrally by the administration of oestrogen.
Collapse
|
64
|
Kaneko T, Kuwaki T, Kashiwadani H. Hypothalamic orexinergic neurons modulate pain and itch in an opposite way: pain relief and itch exacerbation. J Physiol Sci 2022; 72:21. [PMID: 35996084 PMCID: PMC10717118 DOI: 10.1186/s12576-022-00846-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 08/07/2022] [Indexed: 11/10/2022]
Abstract
Pain and itch are recognized as antagonistic sensations; pain suppresses itch and inhibition of pain generates itch. There is still a lack of evidence about the neural mechanism of the interaction between pain and itch in the central nervous system. In this study, we focused on the orexin (ORX) neurons in the lateral hypothalamus (LH), which mediate various "defense responses" when animals confront stressors. We found that the scratching behaviors induced by the pruritogen were significantly suppressed in ORX-neuron-ablated (ORX-abl) mice. The exaggerated pain behavior and attenuated itch behavior observed in ORX-abl mice indicated that ORX neurons modulate pain and itch in an opposite way, i.e., pain relief and itch exacerbation. In addition, most of the ORX neurons responded to both pain and itch input. Our results suggest that ORX neurons inversely regulate pain- and itch-related behaviors, which could be understood as a defense response to cope with stress environment.
Collapse
Affiliation(s)
- Tatsuroh Kaneko
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, 890-8544, Japan
| | - Tomoyuki Kuwaki
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, 890-8544, Japan
| | - Hideki Kashiwadani
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, 890-8544, Japan.
| |
Collapse
|
65
|
Mahringer D, Zmarz P, Okuno H, Bito H, Keller GB. Functional correlates of immediate early gene expression in mouse visual cortex. PEER COMMUNITY JOURNAL 2022; 2:e45. [PMID: 37091727 PMCID: PMC7614465 DOI: 10.24072/pcjournal.156] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
During visual development, response properties of layer 2/3 neurons in visual cortex are shaped by experience. Both visual and visuomotor experience are necessary to co-ordinate the integration of bottom-up visual input and top-down motor-related input. Whether visual and visuomotor experience engage different plasticity mechanisms, possibly associated with the two separate input pathways, is still unclear. To begin addressing this, we measured the expression level of three different immediate early genes (IEG) (c-fos, egr1 or Arc) and neuronal activity in layer 2/3 neurons of visual cortex before and after a mouse's first visual exposure in life, and subsequent visuomotor learning. We found that expression levels of all three IEGs correlated positively with neuronal activity, but that first visual and first visuomotor exposure resulted in differential changes in IEG expression patterns. In addition, IEG expression levels differed depending on whether neurons exhibited primarily visually driven or motor-related activity. Neurons with strong motor-related activity preferentially expressed EGR1, while neurons that developed strong visually driven activity preferentially expressed Arc. Our findings are consistent with the interpretation that bottom-up visual input and top-down motor-related input are associated with different IEG expression patterns and hence possibly also with different plasticity pathways.
Collapse
Affiliation(s)
- David Mahringer
- Faculty of Natural Sciences, University of Basel, Basel, Switzerland
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Pawel Zmarz
- Faculty of Natural Sciences, University of Basel, Basel, Switzerland
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Hiroyuki Okuno
- Department of Biochemistry and Molecular Biology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Kagoshima 890-8544, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Georg B Keller
- Faculty of Natural Sciences, University of Basel, Basel, Switzerland
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| |
Collapse
|
66
|
Weber Boutros S, Unni VK, Raber J. An Adaptive Role for DNA Double-Strand Breaks in Hippocampus-Dependent Learning and Memory. Int J Mol Sci 2022; 23:8352. [PMID: 35955487 PMCID: PMC9368779 DOI: 10.3390/ijms23158352] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 12/10/2022] Open
Abstract
DNA double-strand breaks (DSBs), classified as the most harmful type of DNA damage based on the complexity of repair, lead to apoptosis or tumorigenesis. In aging, DNA damage increases and DNA repair decreases. This is exacerbated in disease, as post-mortem tissue from patients diagnosed with mild cognitive impairment (MCI) or Alzheimer's disease (AD) show increased DSBs. A novel role for DSBs in immediate early gene (IEG) expression, learning, and memory has been suggested. Inducing neuronal activity leads to increases in DSBs and upregulation of IEGs, while increasing DSBs and inhibiting DSB repair impairs long-term memory and alters IEG expression. Consistent with this pattern, mice carrying dominant AD mutations have increased baseline DSBs, and impaired DSB repair is observed. These data suggest an adaptive role for DSBs in the central nervous system and dysregulation of DSBs and/or repair might drive age-related cognitive decline (ACD), MCI, and AD. In this review, we discuss the adaptive role of DSBs in hippocampus-dependent learning, memory, and IEG expression. We summarize IEGs, the history of DSBs, and DSBs in synaptic plasticity, aging, and AD. DSBs likely have adaptive functions in the brain, and even subtle alterations in their formation and repair could alter IEGs, learning, and memory.
Collapse
Affiliation(s)
- Sydney Weber Boutros
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA;
| | - Vivek K. Unni
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA;
- Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, OR 97239, USA
- Oregon Health & Science University Parkinson Center, Portland, OR 97239, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA;
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA;
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR 97239, USA
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| |
Collapse
|
67
|
Kim J, Kang S, Choi TY, Chang KA, Koo JW. Metabotropic Glutamate Receptor 5 in Amygdala Target Neurons Regulates Susceptibility to Chronic Social Stress. Biol Psychiatry 2022; 92:104-115. [PMID: 35314057 DOI: 10.1016/j.biopsych.2022.01.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 01/10/2022] [Accepted: 01/13/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND Metabotropic glutamate receptor 5 (mGluR5) has been implicated in stress-related psychiatric disorders, particularly major depressive disorder. Although growing evidence supports the proresilient role of mGluR5 in corticolimbic circuitry in the depressive-like behaviors following chronic stress exposure, the underlying neural mechanisms, including circuits and molecules, remain unknown. METHODS We measured the c-Fos expression and probability of neurotransmitter release in and from basolateral amygdala (BLA) neurons projecting to the medial prefrontal cortex (mPFC) and to the ventral hippocampus (vHPC) after chronic social defeat stress. The role of BLA projections in depressive-like behaviors was assessed using optogenetic manipulations, and the underlying molecular mechanisms of mGluR5 and downstream signaling were investigated by Western blotting, viral-mediated gene transfer, and pharmacological manipulations. RESULTS Chronic social defeat stress disrupted neural activity and glutamatergic transmission in both BLA projections. Optogenetic activation of BLA projections reversed the detrimental effects of chronic social defeat stress on depressive-like behaviors and mGluR5 expression in the mPFC and vHPC. Conversely, inhibition of BLA projections of mice undergoing subthreshold social defeat stress induced a susceptible phenotype and mGluR5 reduction. These two BLA circuits appeared to act in an independent way. We demonstrate that mGluR5 overexpression in the mPFC or vHPC was proresilient while the mGluR5 knockdown was prosusceptible and that the proresilient effects of mGluR5 are mediated through distinctive downstream signaling pathways in the mPFC and vHPC. CONCLUSIONS These findings identify mGluR5 in the mPFC and vHPC that receive BLA inputs as a critical mediator of stress resilience, highlighting circuit-specific signaling for depressive-like behaviors.
Collapse
Affiliation(s)
- Jeongseop Kim
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Shinwoo Kang
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea; Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Tae-Yong Choi
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Keun-A Chang
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea; Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea.
| | - Ja Wook Koo
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea.
| |
Collapse
|
68
|
Aomine Y, Sakurai K, Macpherson T, Ozawa T, Miyamoto Y, Yoneda Y, Oka M, Hikida T. Importin α3 (KPNA3) Deficiency Augments Effortful Reward-Seeking Behavior in Mice. Front Neurosci 2022; 16:905991. [PMID: 35844217 PMCID: PMC9279672 DOI: 10.3389/fnins.2022.905991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
Importin α3 (Gene: Kpna3, the ortholog of human Importin α4) is a member of the importin α family and participates in nucleocytoplasmic transport by forming trimeric complexes between cargo proteins and importin β1. Evidence from human studies has indicated that single nucleotide polymorphisms (SNP) in the KPNA3 gene are associated with the occurrence of several psychiatric disorders accompanied by abnormal reward-related behavior, including schizophrenia, major depression, and substance addiction. However, the precise roles of importin α3 in controlling reward processing and motivation are still unclear. In this study, we evaluated the behavioral effects of Kpna3 knockout (KO) in mice on performance in touchscreen operant chamber-based tasks evaluating simple (fixed-ratio) and effortful (progressive-ratio) reward-seeking behaviors. While Kpna3 KO mice showed no significant differences in operant reward learning on a fixed-ratio schedule, they demonstrated significantly increased motivation (increased break point) to instrumentally respond for sucrose on a progressive-ratio schedule. We additionally measured the number of c-Fos-positive cells, a marker of neural activity, in 20 regions of the brain and identified a network of brain regions based on their interregional correlation coefficients. Network and graph-theoretic analyses suggested that Kpna3 deficiency enhanced overall interregional functional connectivity. These findings suggest the importance of Kpna3 in motivational control and indicate that Kpna3 KO mice may be an attractive line for modeling motivational abnormalities associated with several psychiatric disorders.
Collapse
Affiliation(s)
- Yoshiatsu Aomine
- Laboratory for Advanced Brain Functions, Institute for Protein Research, Osaka University, Osaka, Japan
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| | - Koki Sakurai
- Laboratory for Advanced Brain Functions, Institute for Protein Research, Osaka University, Osaka, Japan
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| | - Tom Macpherson
- Laboratory for Advanced Brain Functions, Institute for Protein Research, Osaka University, Osaka, Japan
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| | - Takaaki Ozawa
- Laboratory for Advanced Brain Functions, Institute for Protein Research, Osaka University, Osaka, Japan
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| | - Yoichi Miyamoto
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Yoshihiro Yoneda
- National Institutes for Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Masahiro Oka
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Takatoshi Hikida
- Laboratory for Advanced Brain Functions, Institute for Protein Research, Osaka University, Osaka, Japan
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
- *Correspondence: Takatoshi Hikida,
| |
Collapse
|
69
|
Pedroso J, Schneider SE, Lima-Rezende CA, Aguiar GPS, Müller LG, Oliveira JV, Piato A, Siebel AM. Evaluation of Resveratrol and Piceatannol Anticonvulsant Potential in Adult Zebrafish (Danio rerio). Neurochem Res 2022; 47:3250-3260. [PMID: 35750876 DOI: 10.1007/s11064-022-03656-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/10/2022] [Accepted: 06/12/2022] [Indexed: 01/14/2023]
Abstract
Epilepsy is a common neurological disorder which affects 50 million people worldwide. Patients with epilepsy may present cognitive deficits and psychological impairment. Currently, 30% of patients fail to respond to any available antiseizure drug, and a significant number of patients do not well tolerate the offered treatments. Then, it is necessary to find out alternatives for controlling epileptic seizures. Studies have shown that despite its neuroprotective effects, resveratrol shows poor anticonvulsant properties. Resveratrol analog, piceatannol, possesses higher biological activity than resveratrol and could be an alternative to control seizure. Thus, the present study investigated the effects of resveratrol and piceatannol in pentylenetetrazole-induced seizures in adult zebrafish (Danio rerio). Only the experimental positive control (diazepam) showed anticonvulsant effect in this study. In addition, no behavioral changes were observed 24 h after seizure occurrence. Finally, the expression of genes related to neuronal activity (c-fos), neurogenesis (p70S6Ka and p70S6Kb), inflammatory response (interleukin 1β), and cell apoptosis (caspase-3) did not change by pentylenetetrazole-induced seizures. Therefore, we failed to observe any anticonvulsant and neuroprotective potential of resveratrol and piceatannol in adult zebrafish. However, resveratrol and piceatannol benefits in epilepsy are not discharged, and more studies are necessary.
Collapse
Affiliation(s)
- Jefferson Pedroso
- Curso de Ciências Biológicas, Universidade Comunitária da Região de Chapecó, Chapecó, SC, Brazil
| | - Sabrina Ester Schneider
- Curso de Ciências Biológicas, Universidade Comunitária da Região de Chapecó, Chapecó, SC, Brazil
| | - Cássia Alves Lima-Rezende
- División Ornitología, Museo Argentino de Ciencias Naturales, Buenos Aires, Argentina.,Programa de Pós-Graduação em Ciências Ambientais, Universidade Comunitária da Região de Chapecó, Chapecó, SC, Brazil
| | - Gean Pablo S Aguiar
- Programa de Pós-Graduação em Ciências Ambientais, Universidade Comunitária da Região de Chapecó, Chapecó, SC, Brazil
| | - Liz Girardi Müller
- Programa de Pós-Graduação em Ciências Ambientais, Universidade Comunitária da Região de Chapecó, Chapecó, SC, Brazil
| | - J Vladimir Oliveira
- Programa de Pós-Graduação em Ciências Ambientais, Universidade Comunitária da Região de Chapecó, Chapecó, SC, Brazil.,Departamento de Engenharia Química e de Alimentos, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Angelo Piato
- Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Anna Maria Siebel
- Curso de Ciências Biológicas, Universidade Comunitária da Região de Chapecó, Chapecó, SC, Brazil. .,Programa de Pós-Graduação em Ciências Ambientais, Universidade Comunitária da Região de Chapecó, Chapecó, SC, Brazil.
| |
Collapse
|
70
|
Spinieli RL, Ben Musa R, Cornelius-Green J, Hasser EM, Cummings KJ. Orexin facilitates the ventilatory and behavioral responses of rats to hypoxia. Am J Physiol Regul Integr Comp Physiol 2022; 322:R581-R596. [PMID: 35380477 PMCID: PMC9109809 DOI: 10.1152/ajpregu.00334.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/22/2022] [Accepted: 03/30/2022] [Indexed: 02/03/2023]
Abstract
Orexin neurons are sensitive to CO2 and contribute to cardiorespiratory homeostasis as well as sensorimotor control. Whether orexin facilitates respiratory and behavioral responses to acute hypoxia is unclear. We hypothesized that orexin neurons are activated by acute hypoxia and that orexin facilitates the hypoxic ventilatory response (HVR), as well as the arterial blood pressure (ABP) and behavioral (movement) responses to acute hypoxia. We further hypothesized that orexin has greater effects in the active phase of the rat circadian cycle, when orexin neurons have high activity. Using whole body plethysmography with EEG, EMG, and the dual-orexin receptor (OxR) antagonist suvorexant (20 mg/kg ip), we determined the effect of OxR blockade on the respiratory, ABP, and behavioral responses of adult rats to acute, graded hypoxia ([Formula: see text]= 0.15, 0.13, 0.11, and 0.09) and hyperoxic hypercapnia ([Formula: see text]= 0.05; [Formula: see text]= 0.95). OxR blockade had no effect on eupnea. OxR blockade significantly reduced the HVR in both inactive and active phases, with a stronger effect in the active phase. OxR blockade reduced the behavioral response to acute hypoxia in the active phase. The central component of the ventilatory and the ABP responses to hypercapnia were reduced by OxR blockade solely in the inactive phase. In the inactive phase, hypoxia activated ∼10% of orexin neurons in the perifornical hypothalamus. These data suggest that orexin neurons participate in the peripheral chemoreflex to facilitate the ventilatory and behavioral responses to acute hypoxia in rats, particularly in the active phase. Orexin also facilitates central chemoreflex responses to CO2 in the inactive phase.
Collapse
Affiliation(s)
- Richard L Spinieli
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Ruwaida Ben Musa
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Jennifer Cornelius-Green
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Eileen M Hasser
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Kevin J Cummings
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
71
|
Mu R, Tang S, Han X, Wang H, Yuan D, Zhao J, Long Y, Hong H. A cholinergic medial septum input to medial habenula mediates generalization formation and extinction of visual aversion. Cell Rep 2022; 39:110882. [PMID: 35649349 DOI: 10.1016/j.celrep.2022.110882] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 12/07/2021] [Accepted: 05/06/2022] [Indexed: 12/28/2022] Open
Abstract
Generalization of visual aversion is a critical function of the brain that supports survival, but the underlying neurobiological mechanisms are unclear. We establish a rapid generalization procedure for inducing visual aversion by dynamic stripe images. By using fiber photometry, apoptosis, chemogenetic and optogenetic techniques, and behavioral tests, we find that decreased cholinergic neurons' activity in the medial septum (MS) leads to generalization loss of visual aversion. Strikingly, we identify a projection from MS cholinergic neurons to the medial habenula (MHb) and find that inhibition of the MS→MHb cholinergic circuit disrupts aversion-generalization formation while its continuous activation disrupts subsequent extinction. Further studies show that MS→MHb cholinergic projections modulate the generalization of visual aversion possibly via M1 muscarinic acetylcholine receptors (mAChRs) of downstream neurons coreleasing glutamate and acetylcholine. These findings reveal that the MS→MHb cholinergic circuit is a critical node in aversion-generalization formation and extinction and potentially provides insight into the pathogenesis of affective disorders.
Collapse
Affiliation(s)
- Ronghao Mu
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 211198, China
| | - Susu Tang
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaomeng Han
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 211198, China
| | - Hao Wang
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 211198, China
| | - Danhua Yuan
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 211198, China
| | - Jiajia Zhao
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 211198, China
| | - Yan Long
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 211198, China.
| | - Hao Hong
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
72
|
Barykina NV, Karasev MM, Verkhusha VV, Shcherbakova DM. Technologies for large-scale mapping of functional neural circuits active during a user-defined time window. Prog Neurobiol 2022; 216:102290. [PMID: 35654210 DOI: 10.1016/j.pneurobio.2022.102290] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/15/2022] [Accepted: 05/25/2022] [Indexed: 11/25/2022]
Abstract
The mapping of neural circuits activated during behavior down to individual neurons is crucial for decoding how the brain processes information. Technologies allowing activity-dependent labeling of neurons during user-defined restricted time windows are rapidly developing. Precise marking of the time window with light, in addition to chemicals, is now possible. In these technologies, genetically encoded molecules integrate molecular events resulting from neuronal activity with light/drug-dependent events. The outputs are either changes in fluorescence or activation of gene expression. Molecular reporters allow labeling of activated neurons for visualization and cell-type identification. The transcriptional readout also allows further control of activated neuronal populations using optogenetic tools as reporters. Here we review the design of these technologies and discuss their demonstrated applications to reveal previously unknown connections in the mammalian brain. We also consider the strengths and weaknesses of the current approaches and provide a perspective for the future.
Collapse
Affiliation(s)
- Natalia V Barykina
- P.K. Anokhin Institute of Normal Physiology, Moscow 125315, Russia; Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
| | - Maksim M Karasev
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
| | - Vladislav V Verkhusha
- Department of Genetics, and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
| | - Daria M Shcherbakova
- Department of Genetics, and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
73
|
du Plessis KC, Basu S, Rumbell TH, Lucas EK. Sex-Specific Neural Networks of Cued Threat Conditioning: A Pilot Study. Front Syst Neurosci 2022; 16:832484. [PMID: 35656357 PMCID: PMC9152023 DOI: 10.3389/fnsys.2022.832484] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 04/07/2022] [Indexed: 11/28/2022] Open
Abstract
Cued threat conditioning is the most common preclinical model for emotional memory, which is dysregulated in anxiety disorders and post-traumatic stress disorder. Though women are twice as likely as men to develop these disorders, current knowledge of threat conditioning networks was established by studies that excluded female subjects. For unbiased investigation of sex differences in these networks, we quantified the neural activity marker c-fos across 112 brain regions in adult male and female mice after cued threat conditioning compared to naïve controls. We found that trained females engaged prelimbic cortex, lateral amygdala, cortical amygdala, dorsal peduncular cortex, and subparafasicular nucleus more than, and subparaventricular zone less than, trained males. To explore how these sex differences in regional activity impact the global network, we generated interregional cross-correlations of c-fos expression to identify regions that were co-active during conditioning and performed hub analyses to identify regional control centers within each neural network. These exploratory graph theory-derived analyses revealed sex differences in the functional coordination of the threat conditioning network as well as distinct hub regions between trained males and females. Hub identification across multiple networks constructed by sequentially pruning the least reliable connections revealed globus pallidus and ventral lateral septum as the most robust hubs for trained males and females, respectively. While low sample size and lack of non-associative controls are major limitations, these findings provide preliminary evidence of sex differences in the individual circuit components and broader global networks of threat conditioning that may confer female vulnerability to fear-based psychiatric disease.
Collapse
Affiliation(s)
- Kamryn C. du Plessis
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Sreetama Basu
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, United States
| | - Timothy H. Rumbell
- IBM Thomas J. Watson Research Center, Yorktown Heights, NY, United States
| | - Elizabeth K. Lucas
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- *Correspondence: Elizabeth K. Lucas,
| |
Collapse
|
74
|
Jeton F, Perrin-Terrin AS, Yegen CH, Marchant D, Richalet JP, Pichon A, Boncoeur E, Bodineau L, Voituron N. In Transgenic Erythropoietin Deficient Mice, an Increase in Respiratory Response to Hypercapnia Parallels Abnormal Distribution of CO 2/H +-Activated Cells in the Medulla Oblongata. Front Physiol 2022; 13:850418. [PMID: 35514353 PMCID: PMC9061944 DOI: 10.3389/fphys.2022.850418] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/21/2022] [Indexed: 12/03/2022] Open
Abstract
Erythropoietin (Epo) and its receptor are expressed in central respiratory areas. We hypothesized that chronic Epo deficiency alters functioning of central respiratory areas and thus the respiratory adaptation to hypercapnia. The hypercapnic ventilatory response (HcVR) was evaluated by whole body plethysmography in wild type (WT) and Epo deficient (Epo-TAgh) adult male mice under 4%CO2. Epo-TAgh mice showed a larger HcVR than WT mice because of an increase in both respiratory frequency and tidal volume, whereas WT mice only increased their tidal volume. A functional histological approach revealed changes in CO2/H+-activated cells between Epo-TAgh and WT mice. First, Epo-TAgh mice showed a smaller increase under hypercapnia in c-FOS-positive number of cells in the retrotrapezoid nucleus/parafacial respiratory group than WT, and this, independently of changes in the number of PHOX2B-expressing cells. Second, we did not observe in Epo-TAgh mice the hypercapnic increase in c-FOS-positive number of cells in the nucleus of the solitary tract present in WT mice. Finally, whereas hypercapnia did not induce an increase in the c-FOS-positive number of cells in medullary raphe nuclei in WT mice, chronic Epo deficiency leads to raphe pallidus and magnus nuclei activation by hyperacpnia, with a significant part of c-FOS positive cells displaying an immunoreactivity for serotonin in the raphe pallidus nucleus. All of these results suggest that chronic Epo-deficiency affects both the pattern of ventilatory response to hypercapnia and associated medullary respiratory network at adult stage with an increase in the sensitivity of 5-HT and non-5-HT neurons of the raphe medullary nuclei leading to stimulation of fR for moderate level of CO2.
Collapse
Affiliation(s)
- Florine Jeton
- Laboratoire "Hypoxie et Poumons", UMR INSERM U1272, Université Paris 13, UFR SMBH, Bobigny, France.,Laboratory of Excellence (Labex) GR-Ex, PRES Sorbonne Paris Cité, Paris, France
| | - Anne-Sophie Perrin-Terrin
- Laboratoire "Hypoxie et Poumons", UMR INSERM U1272, Université Paris 13, UFR SMBH, Bobigny, France.,Inserm, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Sorbonne Université, Paris, France
| | - Celine-Hivda Yegen
- Laboratoire "Hypoxie et Poumons", UMR INSERM U1272, Université Paris 13, UFR SMBH, Bobigny, France
| | - Dominique Marchant
- Laboratoire "Hypoxie et Poumons", UMR INSERM U1272, Université Paris 13, UFR SMBH, Bobigny, France
| | - Jean-Paul Richalet
- Laboratoire "Hypoxie et Poumons", UMR INSERM U1272, Université Paris 13, UFR SMBH, Bobigny, France.,Laboratory of Excellence (Labex) GR-Ex, PRES Sorbonne Paris Cité, Paris, France
| | - Aurélien Pichon
- Laboratoire "Hypoxie et Poumons", UMR INSERM U1272, Université Paris 13, UFR SMBH, Bobigny, France.,Laboratory of Excellence (Labex) GR-Ex, PRES Sorbonne Paris Cité, Paris, France
| | - Emilie Boncoeur
- Laboratoire "Hypoxie et Poumons", UMR INSERM U1272, Université Paris 13, UFR SMBH, Bobigny, France
| | - Laurence Bodineau
- Inserm, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Sorbonne Université, Paris, France
| | - Nicolas Voituron
- Laboratoire "Hypoxie et Poumons", UMR INSERM U1272, Université Paris 13, UFR SMBH, Bobigny, France.,Laboratory of Excellence (Labex) GR-Ex, PRES Sorbonne Paris Cité, Paris, France
| |
Collapse
|
75
|
Stressed rats fail to exhibit avoidance reactions to innately aversive social calls. Neuropsychopharmacology 2022; 47:1145-1155. [PMID: 34848856 PMCID: PMC9018727 DOI: 10.1038/s41386-021-01230-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 10/01/2021] [Accepted: 10/30/2021] [Indexed: 02/02/2023]
Abstract
Disruptions in amygdalar function, a brain area involved in encoding emotionally salient information, has been implicated in stress-related affective disorders. Earlier animal studies on the behavioral consequences of stress-induced abnormalities in the amygdala focused on learned behaviors using fear conditioning paradigms. If and how stress affects unconditioned, innate fear responses to ethologically natural aversive stimuli remains unexplored. Hence, we subjected rats to aversive ultrasonic vocalization calls emitted on one end of a linear track. Unstressed control rats exhibited a robust avoidance response by spending more time away from the source of the playback calls. Unexpectedly, prior exposure to chronic immobilization stress prevented this avoidance reaction, rather than enhancing it. Further, this stress-induced impairment extended to other innately aversive stimuli, such as white noise and electric shock in an inhibitory avoidance task. However, conditioned fear responses were enhanced by the same stress. Inactivation of the basolateral amygdala (BLA) in control rats prevented this avoidance reaction evoked by the playback. Consistent with this, analysis of the immediate early gene cFos revealed higher activity in the BLA of control, but not stressed rats, after exposure to the playback. Further, in vivo recordings in freely behaving control rats exposed to playback showed enhanced theta activity in the BLA, which also was absent in stressed rats. These findings offer a new framework for studying stress-induced alterations in amygdala-dependent maladaptive responses to more naturally threatening and emotionally relevant social stimuli. The divergent impact of stress on defensive responses--impaired avoidance responses together with increased conditioned fear--also has important implications for models of learned helplessness and depression.
Collapse
|
76
|
Singh T, Batabyal T, Kapur J. Neuronal circuits sustaining neocortical-injury-induced status epilepticus. Neurobiol Dis 2022; 165:105633. [PMID: 35065250 PMCID: PMC8860889 DOI: 10.1016/j.nbd.2022.105633] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 01/04/2022] [Accepted: 01/16/2022] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVES Acute injuries or insults to the cortex, such as trauma, subarachnoid hemorrhage, lobar hemorrhage, can cause seizures or status epilepticus(SE). Neocortical SE is associated with coma, worse prognosis, delayed recovery, and the development of epilepsy. The anatomical structures progressively recruited during neocortical-onset status epilepticus (SE) is unknown. Therefore, we constructed large-scale maps of brain regions active during neocortical SE. METHODS We used a neocortical injury-induced SE mouse model. We implanted cobalt (Co) in the right supplementary motor cortex (M2). We 16 h later administered a homocysteine injection (845 mg/kg, intraperitoneal) to C57Bl/6 J mice to induce SE and monitored it by video and EEG. We harvested animals for 1 h (early-stage) and 2 h (late-stage) following homocysteine injections. To construct activation maps, we immunolabeled whole-brain sections for cFos and NeuN, imaged them using a confocal microscope and quantified cFos immunoreactivity (IR). RESULTS SE in the early phase consisted of discrete, focal intermittent seizures, which became continuous and bilateral in the late stage. In this early stage, cFos IR was primarily observed in the right hemisphere, ipsilateral to the Co lesion, specifically in the motor cortex, retrosplenial cortex, somatosensory cortex, anterior cingulate cortex, lateral and medial septal nuclei, and amygdala. We observed bilateral cFos IR in brain regions during the late stage, indicating the bilateral spread of focal seizures. We found increased cFOS IR in the bilateral somatosensory cortex and the motor cortex and subcortical regions, including the amygdala, thalamus, and hypothalamus. There was noticeably different, intense cFos IR in the bilateral hippocampus compared to the early stage. In addition, there was higher activity in the cortex ipsilateral to the seizure focus during the late stage compared with the early one. CONCLUSION We present a large-scale, high-resolution map of seizure spread during neocortical injury-induced SE. Cortico-cortical and cortico subcortical re-entrant circuits sustain neocortical SE. Neuronal loss following neocortical SE, distant from the neocortical focus, may result from seizures.
Collapse
Affiliation(s)
- Tanveer Singh
- Department of Neurology, University of Virginia, Charlottesville, VA 22908, USA
| | - Tamal Batabyal
- UVA Brain Institute, University of Virginia, Charlottesville, VA 22908, USA
| | - Jaideep Kapur
- Department of Neurology, University of Virginia, Charlottesville, VA 22908, USA; UVA Brain Institute, University of Virginia, Charlottesville, VA 22908, USA; Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
77
|
Chen Z, Ding Y, Zeng Y, Zhang XP, Chen JY. Dexmedetomidine reduces propofol-induced hippocampal neuron injury by modulating the miR-377-5p/Arc pathway. BMC Pharmacol Toxicol 2022; 23:18. [PMID: 35337381 PMCID: PMC8957152 DOI: 10.1186/s40360-022-00555-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 03/08/2022] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Propofol and dexmedetomidine (DEX) are widely used in general anesthesia, and exert toxic and protective effects on hippocampal neurons, respectively. The study sought to investigate the molecular mechanisms of DEX-mediated neuroprotection against propofol-induced hippocampal neuron injury in mouse brains. METHODS Hippocampal neurons of mice and HT22 cells were treated with propofol, DEX, and propofol+DEX. In addition, transfection of miR-377-5p mimics or inhibitors was performed in HT22 cells. Neuronal apoptosis was evaluated by a means of terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) or Hochest 33,258 staining; Arc positive expression in hippocampus tissues was detected using a microscope in immunohistochemistry assays; miRNA-377-5p expression was quantified by RT-qPCR; the protein levels of Arc, DNMT3A, and DNMT3B were determined using western blot; Cell Counting Kit-8 (CCK-8) assay was used to detect the viability and apoptotic rate of the neurons; methylation analysis in the miR-377-5p promoter was performed through methylated DNA immunoprecipitation (MeDIP) assay; dual luciferase reporter assay was performed to confirm whether Arc was under targeted regulation of miR-377-5p. RESULTS In the current study, both in vitro and in vivo, propofol treatment induced hippocampal neuron apoptosis and suppressed cell viability. DNMT3A and DNMT3B expression levels were decreased following propofol treatment, resulting in lowered methylation in the miR-377-5p promoter region and then enhanced expression of miR-377-5p, leading to a decrease in the expression of downstream Arc. Conversely, the expression levels of DNMT3A and DNMT3B were increased following DEX treatment, thus methylation in miR-377-5p promoter region was improved, and miR-377-5p expression was decreased, leading to an increase in the expression of downstream Arc. Eventually, DEX pretreatment protected hippocampal neurons against propofol-induced neurotoxicity by recovering the expression levels of DNMT3A, miR-377-5p, and Arc to the normal levels. Additionally, DNMT3A knockdown improved miR-377-5p expression but reduced Arc expression, and DNMT3A overexpression exerted the opposite effects. Dual luciferase reporter assay revealed a binding target between miR-377-5p and Arc 3'UTR. The neuroprotective effect of DEX against propofol-induced neuronal apoptosis was diminished after Arc knockdown. Silencing Arc independently triggered the apoptosis of HT22 cells, which was alleviated through transfection of miR-377-5p inhibitors. CONCLUSIONS DEX reduced propofol-induced hippocampal neuron injury via the miR-377-5p/Arc signaling pathway.
Collapse
Affiliation(s)
- Zong Chen
- Department of Anesthesiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, NO.19 Nonglin Road, Yuexiu District, Guangzhou, Guangdong Province, China
| | - Yong Ding
- Department of Anesthesiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, NO.19 Nonglin Road, Yuexiu District, Guangzhou, Guangdong Province, China
| | - Ying Zeng
- Department of Anesthesiology, Shenzhen Shajin Hospital Affiliated to Guangzhou Medical University, Shenzhen, China
| | - Xue-Ping Zhang
- Department of Anesthesiology, Shenzhen People's Hospital, Shenzhen Anesthesiology Engineering Center, The Second Clinical Medical College of Jinan University, NO. 1017 Dongmen North Road, Luohu District, Shenzhen, Guangdong Province, China.
| | - Jian-Yan Chen
- Department of Anesthesiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, NO.19 Nonglin Road, Yuexiu District, Guangzhou, Guangdong Province, China.
- Department of Anesthesiology, Shenzhen Shajin Hospital Affiliated to Guangzhou Medical University, Shenzhen, China.
| |
Collapse
|
78
|
Sanada K, Ueno H, Miyamoto T, Baba K, Tanaka K, Nishimura H, Nishimura K, Sonoda S, Yoshimura M, Maruyama T, Onaka T, Otsuji Y, Kataoka M, Ueta Y. AVP-eGFP was significantly upregulated by hypovolemia in the parvocellular division of the paraventricular nucleus in the transgenic rats. Am J Physiol Regul Integr Comp Physiol 2022; 322:R161-R169. [PMID: 35018823 DOI: 10.1152/ajpregu.00107.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 01/06/2022] [Indexed: 11/22/2022]
Abstract
Arginine vasopressin (AVP) is produced in the paraventricular (PVN) and supraoptic nuclei (SON). Peripheral AVP, which is secreted from the posterior pituitary, is produced in the magnocellular division of the PVN (mPVN) and SON. In addition, AVP is produced in the parvocellular division of the PVN (pPVN), where corticotrophin-releasing factor (CRF) is synthesized. These peptides synergistically modulate the hypothalamic-pituitary-adrenal (HPA) axis. Previous studies have revealed that the HPA axis was activated by hypovolemia. However, the detailed dynamics of AVP in the pPVN under hypovolemic state has not been elucidated. Here, we evaluated the effects of hypovolemia and hyperosmolality on the hypothalamus, using AVP-enhanced green fluorescent protein (eGFP) transgenic rats. Polyethylene glycol (PEG) or 3% hypertonic saline (HTN) was intraperitoneally administered to develop hypovolemia or hyperosmolality. AVP-eGFP intensity was robustly upregulated at 3 and 6 h after intraperitoneal administration of PEG or HTN in the mPVN. While in the pPVN, eGFP intensity was significantly increased at 6 h after intraperitoneal administration of PEG with significant induction of Fos-immunoreactive (-ir) neurons. Consistently, eGFP mRNA, AVP hnRNA, and CRF mRNA in the pPVN and plasma AVP and corticosterone were significantly increased at 6 h after intraperitoneal administration of PEG. The results suggest that AVP and CRF syntheses in the pPVN were activated by hypovolemia, resulting in the activation of the HPA axis.
Collapse
Affiliation(s)
- Kenya Sanada
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
- Second Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Hiromichi Ueno
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
- Second Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Tetsu Miyamoto
- Second Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kazuhiko Baba
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kentaro Tanaka
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Haruki Nishimura
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kazuaki Nishimura
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Satomi Sonoda
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Mitsuhiro Yoshimura
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Takashi Maruyama
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Tatsushi Onaka
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Shimotsuke, Japan
| | - Yutaka Otsuji
- Second Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Masaharu Kataoka
- Second Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yoichi Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
79
|
Dutta SS, Andonova AA, Wöllert T, Hewett SJ, Hewett JA. P2X7-dependent constitutive Interleukin-1β release from pyramidal neurons of the normal mouse hippocampus: Evidence for a role in maintenance of the innate seizure threshold. Neurobiol Dis 2022; 168:105689. [DOI: 10.1016/j.nbd.2022.105689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/02/2022] [Accepted: 03/07/2022] [Indexed: 11/30/2022] Open
|
80
|
Finton CJ, Kelly AM, Ophir AG. Support for the parental practice hypothesis: Subadult prairie voles exhibit similar behavioral and neural profiles when alloparenting kin and non-kin. Behav Brain Res 2022; 417:113571. [PMID: 34499932 PMCID: PMC8578440 DOI: 10.1016/j.bbr.2021.113571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 07/09/2021] [Accepted: 08/26/2021] [Indexed: 01/26/2023]
Abstract
Parental care is critical for offspring survival in altricial species. Although parents are the most common caregivers, other individuals (e.g., older siblings) can also provide alloparental care. Some have argued that animals engage in alloparental behavior to practice providing care for their eventual offspring, whereas others have argued that alloparental behavior enhances indirect fitness. Proximate measures have the potential to test ultimate functions of behavior. A focus on neural expression of oxytocin and vasopressin (two neuropeptides modulating alloparental care) or neural activation following exposure to related and unrelated individuals could reveal whether practice or investment in indirect fitness explains alloparental behavior. This study examined alloparental behaviors and neural responses in prairie voles (Microtus ochrogaster), a species that engages in alloparental behavior. Subadult (independent, yet sexually immature) male prairie voles were exposed to one of four stimuli: same-age sibling, neonatal sibling, unrelated neonate, or inanimate neonate-sized object. We assessed alloparental behaviors and quantified cFos protein expression in oxytocin and vasopressin neuronal populations of the paraventricular nucleus of the hypothalamus and the supraoptic nucleus of the hypothalamus in response to stimulus exposure. We detected no differences in cFos and nonapeptide co-localization among stimulus groups. Subjects performed similar amounts of alloparental care toward related and unrelated neonates, but not other subadults or inanimate objects. Notably, caregiving did not differ based on kin-status. The lack of difference in alloparenting toward related and non-related neonates suggests that alloparental care in prairie voles primarily serves to provide subadults with parental practice.
Collapse
Affiliation(s)
| | | | - Alexander G. Ophir
- Department of Psychology, Cornell University, Ithaca, NY,Corresponding author: Alexander Ophir 211 Uris Hall Department of Psychology Cornell University Ithaca, NY 14853, USA Phone: 1-607-255-3714
| |
Collapse
|
81
|
Sebastián-Serrano Á, Merchán-Rubira J, Di-Lauro C, Bianchi C, Soria-Tobar L, Narisawa S, Millán JL, Ávila J, Hernández F, Díaz-Hernández M. TNAP upregulation is a critical factor in Tauopathies and its blockade ameliorates neurotoxicity and increases life-expectancy. Neurobiol Dis 2022; 165:105632. [DOI: 10.1016/j.nbd.2022.105632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 12/22/2022] Open
|
82
|
Nosjean A, Granon S. Brain Adaptation to Acute Stress: Effect of Time, Social Buffering, and Nicotinic Cholinergic System. Cereb Cortex 2021; 32:3990-4011. [PMID: 34905774 DOI: 10.1093/cercor/bhab461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 11/13/2022] Open
Abstract
Both social behavior and stress responses rely on the activity of the prefrontal cortex (PFC) and basolateral nucleus of the amygdala (BLA) and on cholinergic transmission. We previously showed in adult C57BL/6J (B6) mice that social interaction has a buffering effect on stress-related prefrontal activity, depending on the β2-/- cholinergic nicotinic receptors (nAChRs, β2-/- mice). The latency for this buffer to emerge being short, we question here whether the associated brain plasticity, as reflected by regional c-fos protein quantification and PFC-BLA functional connectivity, is modulated by time. Overall, we show that time normalized the stress-induced PFC hyperactivation in B6 mice and PFC hypo-activation in β2-/- mice, with no effect on BLA. It also triggered a multitude of functional links between PFC subareas, and between PFC and BLA in B6 mice but not β2-/- mice, showing a central role of nAChRs in this plasticity. Coupled with social interaction and time, stress led to novel and drastic diminution of functional connectivity within the PFC in both genotypes. Thus, time, emotional state, and social behavior induced dissociated effects on PFC and BLA activity and important cortico-cortical reorganizations. Both activity and plasticity were under the control of the β2-nAChRs.
Collapse
Affiliation(s)
- Anne Nosjean
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay (NeuroPSI), 91400 Saclay, France
| | - Sylvie Granon
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay (NeuroPSI), 91400 Saclay, France
| |
Collapse
|
83
|
Ren Q, Gao D, Mou L, Zhang S, Zhang M, Li N, Sik A, Jin M, Liu K. Anticonvulsant activity of melatonin and its success in ameliorating epileptic comorbidity-like symptoms in zebrafish. Eur J Pharmacol 2021; 912:174589. [PMID: 34699755 DOI: 10.1016/j.ejphar.2021.174589] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 01/08/2023]
Abstract
Epilepsy is one of common neurological disorders, greatly distresses the well-being of the sufferers. Melatonin has been used in clinical anti-epileptic studies, but its effect on epileptic comorbidities is unknown, and the underlying mechanism needs further investigation. Herein, by generating PTZ-induced zebrafish seizure model, we carried out interdisciplinary research using neurobehavioral assays, bioelectrical detection, molecular biology, and network pharmacology to investigate the activity of melatonin as well as its pharmacological mechanisms. We found melatonin suppressed seizure-like behavior by using zebrafish regular locomotor assays. Zebrafish freezing and bursting activity assays revealed the ameliorative effect of melatonin on comorbidity-like symptoms. The preliminary screening results of neurobehavioral assays were further verified by the expression of key genes involved in neuronal activity, neurodevelopment, depression and anxiety, as well as electrical signal recording from the midbrain of zebrafish. Subsequently, network pharmacology was introduced to identify potential targets of melatonin and its pathways. Real-time qPCR and protein-protein interaction (PPI) were conducted to confirm the underlying mechanisms associated with glutathione metabolism. We also found that melatonin receptors were involved in this process, which were regulated in response to melatonin exposure before PTZ treatment. The antagonists of melatonin receptors affected anticonvulsant activity of melatonin. Overall, current study revealed the considerable ameliorative effects of melatonin on seizure and epileptic comorbidity-like symptoms and unveiled the underlying mechanism. This study provides an animal model for the clinical application of melatonin in the treatment of epilepsy and its comorbidities.
Collapse
Affiliation(s)
- Qingyu Ren
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Daili Gao
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Lei Mou
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Shanshan Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Mengqi Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Ning Li
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Attila Sik
- Institute of Physiology, Medical School, University of Pecs, Pecs, H-7624, Hungary; Szentagothai Research Centre, University of Pecs, Pecs, H-7624, Hungary; Institute of Clinical Sciences, Medical School, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China.
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China.
| |
Collapse
|
84
|
Terai H, Gwedela MNV, Kawakami K, Aizawa H. Electrophysiological and pharmacological characterization of spreading depolarization in the adult zebrafish tectum. J Neurophysiol 2021; 126:1934-1942. [PMID: 34731067 DOI: 10.1152/jn.00343.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Spreading depolarization (SD) is a slowly propagating wave of neuronal and glial depolarization. A growing number of studies show that SD and SD-like phenomena play a role in neurological disorders such as migraine, stroke, and traumatic brain injury. Despite the clinical importance of SD, its underlying molecular and cellular mechanisms remain elusive, possibly because of insufficient animal model allowing genetic manipulation. Such a model would also allow high-throughput screening for SD-suppressing drug development. To address this, we developed a novel experimental system to study SD using zebrafish. Electrophysiological recordings in the immobilized adult zebrafish revealed that increasing extracellular potassium concentration elicited SD with a large and long-lasting negative shift of direct current (DC) potential in the optic tectum. It also reduced the oscillatory activity in the extracellular field potential and increased the expression of the immediate early gene c-fos. Pharmacological blocking of the N-methyl-d-aspartate (NMDA) glutamate receptor attenuated the propagation of SD, suggesting that glutamatergic neurotransmission mediated tectal SD in zebrafish. Our analyses revealed that the zebrafish tectum and rodent cortex had similar SD kinetics. The current study provides electrophysiological and pharmacological evidence that zebrafish SD and mammal SD are comparable. This zebrafish SD model is suitable for genetic manipulation and cost-effective high-throughput screening. It could pave the way to novel diagnostic and therapeutic methods applicable to SD-associated neurological disorders.NEW & NOTEWORTHY Previous studies have implicated spreading depolarization (SD) in stroke and migraine. Here, we demonstrate SD, for the first time, in the adult zebrafish tectum showing waveform kinetics, c-fos expression, and attenuation by N-methyl-d-aspartate glutamate receptor blocker as observed in the rodent cortex. Since the zebrafish is an animal model amenable to genetic manipulation and chemical screening, this result could pave the way to novel diagnostic and therapeutic methods applicable to SD-associated neurological disorders.
Collapse
Affiliation(s)
- Haruhi Terai
- Department of Neurobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mayeso Naomi Victoria Gwedela
- Department of Neurobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Koichi Kawakami
- Laboratory of Molecular and Developmental Biology, National Institute of Genetics and Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Shizuoka, Japan
| | - Hidenori Aizawa
- Department of Neurobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
85
|
Matchynski JI, Manwar R, Kratkiewicz KJ, Madangopal R, Lennon VA, Makki KM, Reppen AL, Woznicki AR, Hope BT, Perrine SA, Conti AC, Avanaki K. Direct measurement of neuronal ensemble activity using photoacoustic imaging in the stimulated Fos-LacZ transgenic rat brain: A proof-of-principle study. PHOTOACOUSTICS 2021; 24:100297. [PMID: 34522608 PMCID: PMC8426561 DOI: 10.1016/j.pacs.2021.100297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/28/2021] [Accepted: 08/28/2021] [Indexed: 05/16/2023]
Abstract
Measuring neuroactivity underlying complex behaviors facilitates understanding the microcircuitry that supports these behaviors. We have developed a functional and molecular photoacoustic tomography (F/M-PAT) system which allows direct imaging of Fos-expressing neuronal ensembles in Fos-LacZ transgenic rats with a large field-of-view and high spatial resolution. F/M-PAT measures the beta-galactosidase catalyzed enzymatic product of exogenous chromophore X-gal within ensemble neurons. We used an ex vivo imaging method in the Wistar Fos-LacZ transgenic rat, to detect neuronal ensembles in medial prefrontal cortex (mPFC) following cocaine administration or a shock-tone paired stimulus. Robust and selective F/M-PAT signal was detected in mPFC neurons after both conditions (compare to naive controls) demonstrating successful and direct detection of Fos-expressing neuronal ensembles using this approach. The results of this study indicate that F/M-PAT can be used in conjunction with Fos-LacZ rats to monitor neuronal ensembles that underlie a range of behavioral processes, such as fear learning or addiction.
Collapse
Key Words
- ANSI, American national standards institute
- AP, anterior-posterior
- Brain
- CNR, contrast-to-noise ratio
- Cocaine
- DMSO, dimethyl sulfoxide
- DV, dorsal-ventral
- F/M-PAT, functional molecular photoacoustic tomography
- FOV, field-of-view
- Fear conditioning
- Fos
- GRIN, gradient-index
- IL, infralimbic cortex
- ML, medial-lateral
- Neuronal ensemble
- OCT, optical coherence tomography
- OPO, optical parametric oscillator
- PA, photoacoustic
- PBS, phosphate buffer saline
- PL, prelimbic cortex
- Photoacoustic imaging
- SNR, signal-to-noise ratio
- US, ultrasound
- X-gal
- X-gal, beta-D-galactosidase
- fMRI, functional magnetic resonance imaging
- mPFC, medial prefrontal cortex
Collapse
Affiliation(s)
- James I. Matchynski
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI, USA
- Translational Neuroscience Program, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Rayyan Manwar
- The Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, USA
| | - Karl J. Kratkiewicz
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, USA
| | - Rajtarun Madangopal
- The National Institute on Drug Abuse (NIDA) Intramural Research Program, Baltimore, MD, USA
| | - Veronica A. Lennon
- The National Institute on Drug Abuse (NIDA) Intramural Research Program, Baltimore, MD, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kassem M. Makki
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI, USA
| | - Abbey L. Reppen
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI, USA
| | | | - Bruce T. Hope
- The National Institute on Drug Abuse (NIDA) Intramural Research Program, Baltimore, MD, USA
| | - Shane A. Perrine
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI, USA
- Translational Neuroscience Program, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Alana C. Conti
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI, USA
- Translational Neuroscience Program, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kamran Avanaki
- The Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, USA
- Department of Dermatology, University of Illinois at Chicago, Chicago, USA
| |
Collapse
|
86
|
Mejía-Chávez S, Venebra-Muñoz A, García-García F, Corona-Morales AA, Orozco-Vargas AE. Maternal Separation Modifies the Activity of Social Processing Brain Nuclei Upon Social Novelty Exposure. Front Behav Neurosci 2021; 15:651263. [PMID: 34803620 PMCID: PMC8599987 DOI: 10.3389/fnbeh.2021.651263] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 10/12/2021] [Indexed: 11/13/2022] Open
Abstract
Maternal separation has been shown to disrupt proper brain development and maturation, having profound consequences on the neuroendocrine systems in charge of the stress response, and has been shown to induce behavioral and cognitive abnormalities. At the behavioral level, maternal separation has been shown to increase offensive play-fighting in juvenile individuals and reduce social interest in adulthood. Since most of the studies that have evaluated the consequences of maternal separation on social behavior have focused on behavioral analysis, there is a need for a further understanding of the neuronal mechanisms underlying the changes in social behavior induced by maternal separation. Therefore, the aim of the present research was to assess the long-term effects of maternal separation on social interaction behavior and to assess the activity of several brain regions involved in the processing of social cues and reward upon social novelty exposure, using c-Fos immunohistochemistry as a marker of neuronal activity. Male Wistar rats were subjected to 4 h maternal separation during the neonatal period, 9:00 h-13:00 h from postnatal day 1 to 21, and exposed to social novelty during adulthood. After social novelty exposure, brains were fixed and coronal sections of the medial amygdala, lateral septum (LS), paraventricular nucleus of the hypothalamus, nucleus accumbens, and medial prefrontal cortex were obtained for c-Fos immunohistochemistry. Maternally separated rats spent less time investigating the novel peer, suggesting that maternal separation reduces social approach motivation. Furthermore, maternal separation reduced the number of c-Fos positive cells of the medial amygdala, paraventricular nucleus of the hypothalamus, LS, nucleus accumbens, and medial prefrontal cortex upon social novelty exposure. These findings suggest that maternal separation can reduce the plastic capacity of several brain nuclei, which constitute a physiological basis for the emergence of behavioral disorders presented later in life reported to be linked to early life adversity.
Collapse
Affiliation(s)
- Sara Mejía-Chávez
- Laboratorio de Neurobiología de la Adicción y Plasticidad Cerebral, Facultad de Ciencias, Universidad Autónoma del Estado de Mexico, Toluca, Mexico
| | - Arturo Venebra-Muñoz
- Laboratorio de Neurobiología de la Adicción y Plasticidad Cerebral, Facultad de Ciencias, Universidad Autónoma del Estado de Mexico, Toluca, Mexico
| | - Fabio García-García
- Laboratorio de Biología de Sueño, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
| | | | | |
Collapse
|
87
|
Altered cell and RNA isoform diversity in aging Down syndrome brains. Proc Natl Acad Sci U S A 2021; 118:2114326118. [PMID: 34795060 PMCID: PMC8617492 DOI: 10.1073/pnas.2114326118] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2021] [Indexed: 12/11/2022] Open
Abstract
Down syndrome (DS) neurocognitive disabilities associated with trisomy 21 are known; however, gene changes within individual brain cells occurring with age are unknown. Here, we interrogated >170,000 cells from 29 aging DS and control brains using single-nucleus RNA sequencing. We observed increases in inhibitory-over-excitatory neurons, microglial activation in the youngest DS brains coinciding with overexpression of genes associated with microglial-mediated synaptic pruning, and overexpression of the chromosome 21 gene RUNX1 that may be a potential driving factor in microglial activation. Single-nucleus long-read sequencing revealed hundreds of thousands of unannotated RNA transcripts. These included diverse species for the Alzheimer’s disease gene—amyloid precursor protein—that contained intra-exonic junctions previously associated with somatic gene recombination, which was also identified in ∼8,000 other genes. Down syndrome (DS), trisomy of human chromosome 21 (HSA21), is characterized by lifelong cognitive impairments and the development of the neuropathological hallmarks of Alzheimer’s disease (AD). The cellular and molecular modifications responsible for these effects are not understood. Here we performed single-nucleus RNA sequencing (snRNA-seq) employing both short- (Illumina) and long-read (Pacific Biosciences) sequencing technologies on a total of 29 DS and non-DS control prefrontal cortex samples. In DS, the ratio of inhibitory-to-excitatory neurons was significantly increased, which was not observed in previous reports examining sporadic AD. DS microglial transcriptomes displayed AD-related aging and activation signatures in advance of AD neuropathology, with increased microglial expression of C1q complement genes (associated with dendritic pruning) and the HSA21 transcription factor gene RUNX1. Long-read sequencing detected vast RNA isoform diversity within and among specific cell types, including numerous sequences that differed between DS and control brains. Notably, over 8,000 genes produced RNAs containing intra-exonic junctions, including amyloid precursor protein (APP) that had previously been associated with somatic gene recombination. These and related results illuminate large-scale cellular and transcriptomic alterations as features of the aging DS brain.
Collapse
|
88
|
Mackenzie-Gray Scott CA, Parrish RR, Walsh DA, Racca C, Cowell RM, Treveylan AJ. PV-specific loss of the transcriptional coactivator PGC-1α slows down the evolution of epileptic activity in an acute ictogenic model. J Neurophysiol 2021; 127:86-98. [PMID: 34788174 PMCID: PMC8721902 DOI: 10.1152/jn.00295.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The transcriptional coactivator, PGC-1α (peroxisome proliferator-activated receptor γ coactivator 1α), plays a key role in coordinating energy requirement within cells. Its importance is reflected in the growing number of psychiatric and neurological conditions that have been associated with reduced PGC-1α levels. In cortical networks, PGC-1α is required for the induction of parvalbumin (PV) expression in interneurons, and PGC-1α deficiency affects synchronous GABAergic release. It is unknown, however, how this affects cortical excitability. We show here that knocking down PGC-1α specifically in the PV-expressing cells (PGC-1αPV−/−) blocks the activity-dependent regulation of the synaptic proteins, SYT2 and CPLX1. More surprisingly, this cell class-specific knockout of PGC-1α appears to have a novel antiepileptic effect, as assayed in brain slices bathed in 0 Mg2+ media. The rate of occurrence of preictal discharges developed approximately equivalently in wild-type and PGC-1αPV−/− brain slices, but the intensity of these discharges was lower in PGC-1αPV−/− slices, as evident from the reduced power in the γ range and reduced firing rates in both PV interneurons and pyramidal cells during these discharges. Reflecting this reduced intensity in the preictal discharges, the PGC-1αPV−/− brain slices experienced many more discharges before transitioning into a seizure-like event. Consequently, there was a large increase in the latency to the first seizure-like event in brain slices lacking PGC-1α in PV interneurons. We conclude that knocking down PGC-1α limits the range of PV interneuron firing and this slows the pathophysiological escalation during ictogenesis. NEW & NOTEWORTHY Parvalbumin expressing interneurons are considered to play an important role in regulating cortical activity. We were surprised, therefore, to find that knocking down the transcriptional coactivator, PGC-1α, specifically in this class of interneurons appears to slow ictogenesis. This anti-ictogenic effect is associated with reduced activity in preictal discharges, but with a far longer period of these discharges before the first seizure-like events finally start. Thus, PGC-1α knockdown may promote schizophrenia while reducing epileptic tendencies.
Collapse
Affiliation(s)
| | - Robert Ryley Parrish
- Newcastle University Biosciences Institute, Medical School, Framlington Place, Newcastle upon Tyne, United Kingdom
| | - Darren A Walsh
- Newcastle University Biosciences Institute, Medical School, Framlington Place, Newcastle upon Tyne, United Kingdom
| | - Claudia Racca
- Newcastle University Biosciences Institute, Medical School, Framlington Place, Newcastle upon Tyne, United Kingdom
| | - Rita M Cowell
- Department of Neuroscience, Drug Discovery Division at Southern Research, Birmingham, AL, United States.,Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andrew J Treveylan
- Newcastle University Biosciences Institute, Medical School, Framlington Place, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
89
|
Guo W, Fan S, Xiao D, Dong H, Xu G, Wan Z, Ma Y, Wang Z, Xue T, Zhou Y, Li Y, Xiong W. A Brainstem reticulotegmental neural ensemble drives acoustic startle reflexes. Nat Commun 2021; 12:6403. [PMID: 34737329 PMCID: PMC8568936 DOI: 10.1038/s41467-021-26723-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 10/20/2021] [Indexed: 11/21/2022] Open
Abstract
The reticulotegmental nucleus (RtTg) has long been recognized as a crucial component of brainstem reticular formation (RF). However, the function of RtTg and its related circuits remain elusive. Here, we report a role of the RtTg in startle reflex, a highly conserved innate defensive behaviour. Optogenetic activation of RtTg neurons evokes robust startle responses in mice. The glutamatergic neurons in the RtTg are significantly activated during acoustic startle reflexes (ASR). Chemogenetic inhibition of the RtTg glutamatergic neurons decreases the ASR amplitudes. Viral tracing reveals an ASR neural circuit that the cochlear nucleus carrying auditory information sends direct excitatory innervations to the RtTg glutamatergic neurons, which in turn project to spinal motor neurons. Together, our findings describe a functional role of RtTg and its related neural circuit in startle reflexes, and demonstrate how the RF connects auditory system with motor functions.
Collapse
Affiliation(s)
- Weiwei Guo
- grid.59053.3a0000000121679639Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230026 China
| | - Sijia Fan
- grid.59053.3a0000000121679639Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230026 China
| | - Dan Xiao
- grid.59053.3a0000000121679639Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230026 China
| | - Hui Dong
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, 100871 China
| | - Guangwei Xu
- grid.59053.3a0000000121679639Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230026 China
| | - Zhikun Wan
- grid.59053.3a0000000121679639Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230026 China
| | - Yuqian Ma
- grid.59053.3a0000000121679639Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230026 China
| | - Zhen Wang
- grid.16821.3c0000 0004 0368 8293Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Tian Xue
- grid.59053.3a0000000121679639Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230026 China ,grid.9227.e0000000119573309Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Yifeng Zhou
- grid.59053.3a0000000121679639Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230026 China
| | - Yulong Li
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, 100871 China ,grid.11135.370000 0001 2256 9319PKU-IDG–McGovern Institute for Brain Research, Beijing, 100871 China
| | - Wei Xiong
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230026, China. .,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
90
|
Ueta Y. Transgenic approaches to opening up new fields of vasopressin and oxytocin research. J Neuroendocrinol 2021; 33:e13055. [PMID: 34713515 DOI: 10.1111/jne.13055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 10/07/2021] [Accepted: 10/07/2021] [Indexed: 11/29/2022]
Abstract
Transgenic approaches have been applied to generate transgenic rats that express exogenous genes in arginine vasopressin (AVP)- and oxytocin (OXT)-producing magnocellular neurosecretory cells (MNCs) of the hypothalamic-neurohypophyseal system (HNS). First, the fusion gene that expresses AVP-enhanced green fluorescent protein (eGFP) and OXT-monomeric red fluorescent protein 1 (mRFP1) was used to visualize AVP- and OXT-producing MNCs and their axon terminals in the HNS under fluorescence microscopy. Second, the fusion gene that expresses c-fos-eGFP and c-fos-mRFP1 was used to identify activated neurons physiologically in the central nervous system, including MNCs, circumventricular organs and spinal cord. In addition, AVP-eGFP x c-fos-mRFP1 and OXT-mRFP1 × c-fos-eGFP double transgenic rats were generated to identify activated AVP- and OXT-producing MNCs using appropriate physiological stimuli. Third, the fusion gene that expresses AVP-chanelrhodopsin 2 (ChR2)-eGFP and AVP-hM3Dq-mCherry was used to activate AVP- and OXT-producing MNCs by optogenetic and chemogenetic approaches. In each step, these transgenic approaches in rats have provided new insights on the physiological roles of AVP and OXT not only in the HNS, but also in the whole body. In this review, we summarize the transgenic rats that we generated, as well as related physiological findings.
Collapse
Affiliation(s)
- Yoichi Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
91
|
Petrisko TJ, Konat GW. Peripheral viral challenge increases c-fos level in cerebral neurons. Metab Brain Dis 2021; 36:1995-2002. [PMID: 34406561 DOI: 10.1007/s11011-021-00819-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 08/05/2021] [Indexed: 12/01/2022]
Abstract
Peripheral viral infection can substantially alter brain function. We have previously shown that intraperitoneal (i.p.) injection of a viral mimetic, polyinosinic-polycytidylic acid (PIC), engenders hyperexcitability of cerebral neurons. Because neuronal activity is invariably associated with their expression of the Cfos gene, the present study was undertaken to determine whether PIC challenge also increases neuronal c-fos protein level. Female C57BL/6 mice were i.p. injected with PIC, and neuronal c-fos was analyzed in the motor cortex by immunohistochemistry. PIC challenge instigated a robust increase in the number of c-fos-positive neurons. This increase reached approximately tenfold over control at 24 h. Also, the c-fos staining intensity of individual neurons increased. AMG-487, a specific inhibitor of the chemokine receptor CXCR3, profoundly attenuated the accumulation of neuronal c-fos, indicating the activation of CXCL10/CXCR3 axis as the trigger of the process. Together, these results show that the accumulation of c-fos is a viable readout to assess the response of cerebral neurons to peripheral PIC challenge, and to elucidate the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Tiffany J Petrisko
- Department of Biochemistry, Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Gregory W Konat
- Department of Biochemistry, Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV, 26506, USA.
- Department of Biochemistry, West Virginia University School of Medicine, 4052 HSCN, P.O. Box 9128, Morgantown, WV, 26506-9128, USA.
| |
Collapse
|
92
|
Chung H, Parkhurst CN, Magee EM, Phillips D, Habibi E, Chen F, Yeung BZ, Waldman J, Artis D, Regev A. Joint single-cell measurements of nuclear proteins and RNA in vivo. Nat Methods 2021; 18:1204-1212. [PMID: 34608310 PMCID: PMC8532076 DOI: 10.1038/s41592-021-01278-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 08/19/2021] [Indexed: 02/08/2023]
Abstract
Identifying gene-regulatory targets of nuclear proteins in tissues is a challenge. Here we describe intranuclear cellular indexing of transcriptomes and epitopes (inCITE-seq), a scalable method that measures multiplexed intranuclear protein levels and the transcriptome in parallel across thousands of nuclei, enabling joint analysis of transcription factor (TF) levels and gene expression in vivo. We apply inCITE-seq to characterize cell state-related changes upon pharmacological induction of neuronal activity in the mouse brain. Modeling gene expression as a linear combination of quantitative protein levels revealed genome-wide associations of each TF and recovered known gene targets. TF-associated genes were coexpressed as distinct modules that each reflected positive or negative TF levels, showing that our approach can disentangle relative putative contributions of TFs to gene expression and add interpretability to inferred gene networks. inCITE-seq can illuminate how combinations of nuclear proteins shape gene expression in native tissue contexts, with direct applications to solid or frozen tissues and clinical specimens.
Collapse
Affiliation(s)
- Hattie Chung
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| | - Christopher N Parkhurst
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Emma M Magee
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Devan Phillips
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Genentech, South San Francisco, CA, USA
| | - Ehsan Habibi
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Fei Chen
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | | | - Julia Waldman
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Friedman Center for Nutrition and Inflammation, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Genentech, South San Francisco, CA, USA.
| |
Collapse
|
93
|
Vagus nerve stimulation activates two distinct neuroimmune circuits converging in the spleen to protect mice from kidney injury. Proc Natl Acad Sci U S A 2021; 118:2021758118. [PMID: 33737395 DOI: 10.1073/pnas.2021758118] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Acute kidney injury is highly prevalent and associated with high morbidity and mortality, and there are no approved drugs for its prevention and treatment. Vagus nerve stimulation (VNS) alleviates inflammatory diseases including kidney disease; however, neural circuits involved in VNS-induced tissue protection remain poorly understood. The vagus nerve, a heterogeneous group of neural fibers, innervates numerous organs. VNS broadly stimulates these fibers without specificity. We used optogenetics to selectively stimulate vagus efferent or afferent fibers. Anterograde efferent fiber stimulation or anterograde (centripetal) sensory afferent fiber stimulation both conferred kidney protection from ischemia-reperfusion injury. We identified the C1 neurons-sympathetic nervous system-splenic nerve-spleen-kidney axis as the downstream pathway of vagus afferent fiber stimulation. Our study provides a map of the neural circuits important for kidney protection induced by VNS, which is critical for the safe and effective clinical application of VNS for protection from acute kidney injury.
Collapse
|
94
|
Figueroa C, Yang H, DiSpirito J, Bourgeois JR, Kalyanasundaram G, Doshi I, Bilbo SD, Kopec AM. Morphine exposure alters Fos expression in a sex-, age-, and brain region-specific manner during adolescence. Dev Psychobiol 2021; 63:e22186. [PMID: 34423851 DOI: 10.1002/dev.22186] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/17/2021] [Accepted: 08/08/2021] [Indexed: 11/06/2022]
Abstract
Data in both humans and preclinical animal models clearly indicate drug exposure during adolescence, when the "reward" circuitry of the brain develops, increases the risk of substance use and other mental health disorders later in life. Human data indicate that different neural and behavioral sequelae can be observed in early versus late adolescence. However, most studies with rodent models examine a single adolescent age compared to a mature adult age, and often only in males. Herein, we sought to determine whether the acute response to the opioid morphine would also differ across adolescence, and by sex. By quantifying Fos positive cells, a proxy for neural activity, at different stages during adolescence (pre-, early, mid-, and late adolescence) and in multiple reward regions (prefrontal cortex, nucleus accumbens, caudate/putamen), we determined that the neural response to acute morphine is highly dependent on adolescent age, sex, and brain region. These data suggest that heterogeneity in the consequences of adolescent opioid exposure may be due to age- and sex-specific developmental profiles in individual reward processing regions. In future studies, it will be important to add age within adolescence as an independent variable for a holistic view of healthy or abnormal reward-related neural development.
Collapse
Affiliation(s)
- C Figueroa
- Deptartment of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - H Yang
- Deptartment of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA.,Northeastern University, Boston, MA, USA
| | - J DiSpirito
- Deptartment of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA.,Rensselaer Polytechnic Institute, Troy, NY, USA
| | - J R Bourgeois
- Deptartment of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - G Kalyanasundaram
- Deptartment of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - I Doshi
- Deptartment of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - S D Bilbo
- Deptartment of Psychology and Neuroscience, Duke University, Durham, NC, USA.,Deptartment of Pediatrics, Massachusetts General Hospital, Boston, MA, USA.,Lurie Center for Autism, Harvard Medical School, Boston, MA, USA
| | - A M Kopec
- Deptartment of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA.,Deptartment of Pediatrics, Massachusetts General Hospital, Boston, MA, USA.,Lurie Center for Autism, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
95
|
Bryan de la Peña J, Kunder N, Lou TF, Chase R, Stanowick A, Barragan-Iglesias P, Pancrazio JJ, Campbell ZT. A Role for Translational Regulation by S6 Kinase and a Downstream Target in Inflammatory Pain. Br J Pharmacol 2021; 178:4675-4690. [PMID: 34355805 DOI: 10.1111/bph.15646] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Translational controls pervade neurobiology. Nociceptors play an integral role in the detection and propagation of pain signals. Nociceptors can undergo persistent changes in their intrinsic excitability. Pharmacologic disruption of nascent protein synthesis diminishes acute and chronic forms of pain-associated behaviors. Yet, the targets of translational controls that facilitate plasticity in nociceptors are unclear. EXPERIMENTAL APPROACH We used ribosome profiling to probe the translational landscape in DRG neurons after treatment of the inflammatory mediators NGF and IL-6. We validated the expression dynamics of c-Fos using immunoblotting and immunohistochemistry. Given that inflammation is known to stimulate mTOR signaling, we reasoned that downstream factors (e.g., ribosomal protein S6 kinase 1, S6K1) might control c-Fos levels. We utilized small-molecule inhibitors of S6K1 (DG2) or c-Fos (T-5224) to probe their effects on nociceptor activity in vitro using multi-electrode arrays (MEAs) and pain behavior in vivo using a hyperalgesic priming model. KEY RESULTS We demonstrate that c-Fos is expressed in sensory neurons. Inflammatory mediators that promote pain in both humans and rodents promote c-Fos translation. We demonstrate that the mTOR effector S6K1 is essential for c-Fos biosynthesis. Inhibition of S6K1 or c-Fos with small molecules diminish mechanical and thermal hypersensitivity in response to inflammatory cues. Additionally, both inhibitors reduce evoked nociceptor activity. CONCLUSION Our data reveal a novel role of S6K1 in modulating rapid response to inflammatory mediators, with c-Fos being one key downstream target. Targeting the S6 kinase pathway or c-Fos is an exciting new avenue for pain-modulating compounds.
Collapse
Affiliation(s)
- June Bryan de la Peña
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Nikesh Kunder
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Tzu-Fang Lou
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Rebecca Chase
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Alexander Stanowick
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Paulino Barragan-Iglesias
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA.,Department of Physiology and Pharmacology, Center for Basic Sciences, Autonomous University of Aguascalientes, Aguascalientes, Mexico
| | - Joseph J Pancrazio
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA.,Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Zachary T Campbell
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA.,Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA.,Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
96
|
Tilelli CQ, Flôres LR, Cota VR, Castro OWD, Garcia-Cairasco N. Amygdaloid complex anatomopathological findings in animal models of status epilepticus. Epilepsy Behav 2021; 121:106831. [PMID: 31864944 DOI: 10.1016/j.yebeh.2019.106831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 11/15/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022]
Abstract
Temporal lobe epileptic seizures are one of the most common and well-characterized types of epilepsies. The current knowledge on the pathology of temporal lobe epilepsy relies strongly on studies of epileptogenesis caused by experimentally induced status epilepticus (SE). Although several temporal lobe structures have been implicated in the epileptogenic process, the hippocampal formation is the temporal lobe structure studied in the greatest amount and detail. However, studies in human patients and animal models of temporal lobe epilepsy indicate that the amygdaloid complex can be also an important seizure generator, and several pathological processes have been shown in the amygdala during epileptogenesis. Therefore, in the present review, we systematically selected, organized, described, and analyzed the current knowledge on anatomopathological data associated with the amygdaloid complex during SE-induced epileptogenesis. Amygdaloid complex participation in the epileptogenic process is evidenced, among others, by alterations in energy metabolism, circulatory, and fluid regulation, neurotransmission, immediate early genes expression, tissue damage, cell suffering, inflammation, and neuroprotection. We conclude that major efforts should be made in order to include the amygdaloid complex as an important target area for evaluation in future research on SE-induced epileptogenesis. This article is part of the Special Issue "NEWroscience 2018".
Collapse
Affiliation(s)
- Cristiane Queixa Tilelli
- Laboratory of Physiology, Campus Centro-Oeste Dona Lindu, Universidade Federal de São João del-Rei, Av. Sebastião Gonçalves Coelho, 400, Bairro Belvedere, Divinópolis, MG 35.501-296, Brazil.
| | - Larissa Ribeiro Flôres
- Laboratory of Physiology, Campus Centro-Oeste Dona Lindu, Universidade Federal de São João del-Rei, Av. Sebastião Gonçalves Coelho, 400, Bairro Belvedere, Divinópolis, MG 35.501-296, Brazil
| | - Vinicius Rosa Cota
- Laboratory of Neuroengineering and Neuroscience (LINNce), Department of Electrical Engineering, Campus Santo Antônio, Universidade Federal de São João del-Rei, Praça Frei Orlando, 170, Centro, São João Del Rei, MG 36307-352, Brazil
| | - Olagide Wagner de Castro
- Institute of Biological Sciences and Health, Campus A. C. Simões, Universidade Federal de Alagoas, Av. Lourival Melo Mota, s/n, Tabuleiro do Martins, Maceió, AL 57072-970, Brazil
| | - Norberto Garcia-Cairasco
- Neurophysiology and Experimental Neuroethology Laboratory (LNNE), Department of Physiology, School of Medicine, Universidade de São Paulo, Av. Bandeirantes, 3900, Monte Alegre, Ribeirão Preto, SP 14049-900, Brazil.
| |
Collapse
|
97
|
Fry WM, Ferguson AV. The subfornical organ and organum vasculosum of the lamina terminalis: Critical roles in cardiovascular regulation and the control of fluid balance. HANDBOOK OF CLINICAL NEUROLOGY 2021; 180:203-215. [PMID: 34225930 DOI: 10.1016/b978-0-12-820107-7.00013-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
In this chapter, we review the extensive literature describing the roles of the subfornical organ (SFO), the organum vasculosum of the terminalis (OVLT), and the median preoptic nucleus (MnPO), comprising the lamina terminalis, in cardiovascular regulation and the control of fluid balance. We present this information in the context of both historical and technological developments which can effectively be overlaid upon each other. We describe intrinsic anatomy and connectivity and then discuss early work which described how circulating angiotensin II acts at the SFO to stimulate drinking and increase blood pressure. Extensive studies using direct administration and lesion approaches to highlight the roles of all regions of the lamina terminalis are then discussed. At the cellular level we describe c-Fos and electrophysiological work, which has highlighted an extensive group of circulating hormones which appear to influence the activity of specific neurons in the SFO, OVLT, and MnPO. We highlight optogenetic studies that have begun to unravel the complexities of circuitries underlying physiological outcomes, especially those related to different components of drinking. Finally, we describe the somewhat limited human literature supporting conclusions that these structures play similar and potentially important roles in human physiology.
Collapse
Affiliation(s)
- W Mark Fry
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Alastair V Ferguson
- Department of Biomedical and Molecular Sciences and Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
98
|
Dang J, Paudel YN, Yang X, Ren Q, Zhang S, Ji X, Liu K, Jin M. Schaftoside Suppresses Pentylenetetrazol-Induced Seizures in Zebrafish via Suppressing Apoptosis, Modulating Inflammation, and Oxidative Stress. ACS Chem Neurosci 2021; 12:2542-2552. [PMID: 34128378 DOI: 10.1021/acschemneuro.1c00314] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The lack of disease-modifying therapeutic strategies against epileptic seizures has caused a surge in preclinical research focused on exploring and developing novel therapeutic candidates for epilepsy. Compounds from traditional Chinese medicines (TCMs) have gained much attention for a plethora of neurological diseases, including epilepsy. Herein, for the first time, we evaluated the anticonvulsive effects of schaftoside (SS), a TCM, on pentylenetetrazol (PTZ)-induced epileptic seizures in zebrafish and examined the underlying mechanisms. We observed that SS pretreatments significantly suppressed seizure-like behavior and prolonged the onset of seizures. Zebrafish larvae pretreated with SS demonstrated downregulation of c-fos expression during seizures. PTZ-induced upregulation of apoptotic cells was decreased upon pretreatment with SS. Inflammatory phenomena during seizure progression including the upregulation of interleukin 6 (IL-6), interleukin 1 beta (IL-1β), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) were downregulated upon pretreatment with SS. The PTZ-induced recruitment of immunocytes was in turn reduced upon SS pretreatment. Moreover, SS pretreatment modulated oxidative stress, as demonstrated by decreased levels of catalase (CAT) and increased levels of glutathione peroxidase-1a (GPx1a) and manganese superoxide dismutase (Mn-SOD). However, pretreatment with SS modulated the PTZ-induced downregulation of the relative enzyme activity of CAT, GPx, and SOD. Hence, our findings suggest that SS pretreatment ameliorates PTZ-induced seizures, suppresses apoptosis, and downregulates the inflammatory response and oxidative stress, which potentially protect against further seizures in zebrafish.
Collapse
Affiliation(s)
- Jiao Dang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji’nan 250103, Shandong Province, People’s Republic of China
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji’nan 250103, Shandong Province, People’s Republic of China
| | - Yam Nath Paudel
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Xueliang Yang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji’nan 250103, Shandong Province, People’s Republic of China
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji’nan 250103, Shandong Province, People’s Republic of China
| | - Qingyu Ren
- School of Psychology, North China University of Science and Technology, 21 Bohai Road, Tang’shan 063210, Hebei Province, People’s Republic of China
| | - Shanshan Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji’nan 250103, Shandong Province, People’s Republic of China
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji’nan 250103, Shandong Province, People’s Republic of China
| | - Xiuna Ji
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji’nan 250103, Shandong Province, People’s Republic of China
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji’nan 250103, Shandong Province, People’s Republic of China
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji’nan 250103, Shandong Province, People’s Republic of China
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji’nan 250103, Shandong Province, People’s Republic of China
| | - Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji’nan 250103, Shandong Province, People’s Republic of China
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji’nan 250103, Shandong Province, People’s Republic of China
| |
Collapse
|
99
|
Meenakshi P, Kumar S, Balaji J. In vivo imaging of immediate early gene expression dynamics segregates neuronal ensemble of memories of dual events. Mol Brain 2021; 14:102. [PMID: 34187543 PMCID: PMC8243579 DOI: 10.1186/s13041-021-00798-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/27/2021] [Indexed: 11/10/2022] Open
Abstract
Identification of neurons undergoing plasticity in response to external stimuli is one of the pertinent problems in neuroscience. Immediate early genes (IEGs) are widely used as a marker for neuronal plasticity. Here, we model the dynamics of IEG expression as a consecutive, irreversible first-order reaction with a limiting substrate. First, we develop an analytical framework to show that such a model, together with two-photon in vivo imaging of IEG expression, can be used to identify distinct neuronal subsets representing multiple memories. Using the above combination, we show that the expression kinetics, rather than intensity threshold, can be used to identify neuronal ensembles responding to the presentation of two events in vivo. The analytical expression allowed us to segregate the neurons based on their temporal response to one specific behavioural event, thereby improving the ability to detect plasticity related neurons. We image the retrosplenial cortex (RSc) of cfos-GFP transgenic mice to follow the dynamics of cellular changes resulting from contextual fear conditioning behaviour, enabling us to establish a representation of context in RSc at the cellular scale following memory acquisition. Thus, we obtain a general method that distinguishes neurons that took part in multiple temporally separated events by measuring fluorescence of individual neurons in live mice.
Collapse
Affiliation(s)
- P Meenakshi
- Centre for Neurosciences, Indian Institute of Science, Bangalore, 560012, India
| | - S Kumar
- Centre for Neurosciences, Indian Institute of Science, Bangalore, 560012, India
| | - J Balaji
- Centre for Neurosciences, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
100
|
Sanada K, Yoshimura M, Ikeda N, Baba K, Nishimura H, Nishimura K, Nonaka Y, Maruyama T, Miyamoto T, Mori M, Conway-Campbell B, Lightman S, Kataoka M, Ueta Y. Chemogenetic activation of endogenous arginine vasopressin exerts anorexigenic effects via central nesfatin-1/NucB2 pathway. J Physiol Sci 2021; 71:18. [PMID: 34134629 PMCID: PMC10717637 DOI: 10.1186/s12576-021-00802-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/04/2021] [Indexed: 11/10/2022]
Abstract
We examined whether the chemogenetic activation of endogenous arginine vasopressin (AVP) affects central nesfatin-1/NucB2 neurons, using a transgenic rat line that was previously generated. Saline (1 mL/kg) or clozapine-N-oxide (CNO, 1 mg/mL/kg), an agonist for hM3Dq, was subcutaneously administered in adult male AVP-hM3Dq-mCherry transgenic rats (300-370 g). Food and water intake were significantly suppressed after subcutaneous (s.c.) injection of CNO, with aberrant circadian rhythmicity. The percentages of Fos expression in nesfatin-1/NucB2-immunoreactive neurons were significantly increased in the hypothalamus and brainstem at 120 min after s.c. injection of CNO. Suppressed food intake that was induced by chemogenetic activation of endogenous AVP was ablated after intracerebroventricularly administered nesfatin-1/NucB2-neutralizing antibody in comparison with vehicle, without any alteration of water intake nor circadian rhythmicity. These results suggest that chemogenetic activation of endogenous AVP affects, at least in part, central nesfatin-1/NucB2 neurons and may exert anorexigenic effects in the transgenic rats.
Collapse
Affiliation(s)
- Kenya Sanada
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
- Second Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, 807-8555, Japan
| | - Mitsuhiro Yoshimura
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan.
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS1 3NY, UK.
| | - Naofumi Ikeda
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, 807-8555, Japan
| | - Kazuhiko Baba
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, 807-8555, Japan
| | - Haruki Nishimura
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, 807-8555, Japan
| | - Kazuaki Nishimura
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Yuki Nonaka
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Takashi Maruyama
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Tetsu Miyamoto
- Second Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, 807-8555, Japan
| | - Masatomo Mori
- Research Institute for Metabolism and Obesity, Maebashi, 371-0049, Japan
| | - Becky Conway-Campbell
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS1 3NY, UK
| | - Stafford Lightman
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS1 3NY, UK
| | - Masaharu Kataoka
- Second Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, 807-8555, Japan
| | - Yoichi Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan.
| |
Collapse
|